Title

Feasibility Study in Subjects With Mild to Moderate Alzheimer's Disease
Phase 2a Feasibility Study of T3D-959 in Subjects With Mild to Moderate Alzheimer's Disease
  • Phase

    Phase 1/Phase 2
  • Study Type

    Interventional
  • Intervention/Treatment

    t3d-959 ...
  • Study Participants

    36
The study is a randomized, parallel, 4-dose design in subjects with mild-to-moderate Alzheimer's Disease. Subjects will be randomized to one of 4 doses of T3D-959. Subjects will be evaluated for changes from baseline in cerebral metabolic rate of glucose (FDG-PET imaging), functional connectivity of the hippocampus (BOLD-fMRI), and cognitive function (ADAS-Cog11 and DSST) as well as assessed for safety and tolerability to T3D-959.

An expanded access extension is planed to provide access to study medication to subjects who have completed the main study and requested continued use.
T3D-959 is an orally-delivered, once-a-day administered, small molecule dual nuclear receptor agonist that has been shown in animal and Phase 1 studies in normal human subjects to be safe and well tolerated. The purpose of this clinical study in AD patients is to demonstrate mechanistic proof of concept that T3D-959, can produce desired changes in cerebral glucose metabolism and functional connectivity that may indicate potential for cognitive improvement. The therapeutic approach to be tested is based on two suppositions; (A) ameliorating multiple pathologies in the disease with a single therapy may provide a superior clinical benefit than therapeutic approaches which target a single pathology and (B) correcting insulin resistance in the brain, (highly correlated with AD and potential key driver of AD pathophysiology) and peripherally may be disease remedial.

The brain requires integral insulin signaling for metabolic homeostasis and neuronal plasticity. Insulin resistance disrupts energy balance and signaling networks needed for a broad range of functions. Impaired insulin signaling in neurons enhances apoptosis, promotes oxidative cell death induced by Abeta1-42, increases secretion of Abeta1-42, blocks removal of extracellular Abeta oligomers and increases plaque loads. A growing body of evidence suggests that brain insulin resistance promotes or possibly is the trigger of key pathologies in AD and is supported by observed changes in levels of insulin signaling molecules in AD forebrains and associated changes in memory. Pre-clinical studies in animals have demonstrated the insulin sensitizing activity of T3D-959 and ability to improve multiple pathologies of AD in a rat model of disease.

This non-placebo controlled trial will be conducted in one to three clinical centers. Thirty six (36) patients with mild-to-moderate Alzheimer's disease will be randomly assigned to once daily, orally administered treatment with 3mg, 10mg, 30mg or 90mg doses of T3D-959. Participants will be treated for two weeks and will undergo at baseline and at two weeks; FDG-PET scans to measure brain glucose metabolism, BOLD fMRI scans to measure functional connectivity of the hippocampus, venous blood draws for biomarker analysis and ApoE genotyping, and ADAS-Cog11 and DSST cognitive testing. For monitoring potential toxicities of the drug subjects will undergo physical examination, neurological examination, adverse event review, blood chemistries, and pharmacokinetic (PK) analyses for T3D-959 plasma levels.

BOLD fMRI definition of terms:

GoF (Goodness of Fit): The degree to which the spatial extent and magnitude of one subject's default mode network (DMN) regions matches the one of an elderly control group.

Hippo-PreC Link (Hippocampus - Precuneus Link): The resting-state BOLD signal correlation strength between hippocampus and precuneus regions of interest.

GlobEff_DMN: (Global Efficiency from DMN Regions): The global efficiency among 11 pre-defined default mode network regions.

GlobEff_AAL: (Global Efficiency from AAL Regions): The global efficiency among 90 pre-defined cerebral regions based on automated anatomical labeling.

ALFF_lPCC_PreC: Amplitude of Low Frequency Fluctuations (ALFF) from left posterior cingulate cortex (PCC) and precuneus (PreC).

ALFF_rPCC_PreC: ALFF from right PCC and precuneus. fALFF_lPCC_PreC: Ratio ALFF from left PCC and precuneus. fALFF_rPCC_PreC: Ratio ALFF from right PCC and precuneus. ReHo_lPCC_PreC: Regional Homogeneity in left PCC and PreC. ReHo_rPCC_PreC: Regional Homogeneity in right PCC and PreC. ALFF_lIPL: ALFF from left inferior parietal lobule (IPL). ALFF_rIPL: ALFF from right IPL. fALFF_lIPL: Ratio ALFF from left IPL. fALFF_rIPL : Ratio ALFF from right IPL. ReHo_lIPL: Regional Homogeneity in left IPL. ReHo_rIPL: Regional Homogeneity in right IPL.

Expanded Access Extension: This is an open label 10 visit extension for up to 5 subjects who have completed the T3D959-201 protocol and whose caregivers and physician requested their continued treatment in an expanded access protocol. All subjects enrolled in this study will be treated with a 15mg q.d. dose of T3D959 for six months, regardless of their assigned dose level from the main study. A continued risk/benefit assessment by the investigator will be conducted at each visit to determine the need for treatment continuation. Subjects will be assessed for safety and tolerability to T3D-959 and evaluated for changes from baseline cognitive function via ADAS-Cog11 and DSST testing and global change via CIBIC-plus testing.
Study Started
Jul 31
2015
Primary Completion
May 30
2016
Study Completion
Jun 30
2016
Results Posted
Jul 30
2018
Last Update
Jul 30
2018

Drug T3D-959

The 3mg dosage is supplied as 1mg capsules (three capsules, taken once daily by mouth) The 10mg dosage is supplied as 5mg capsules (two capsules, taken once daily by mouth) The 30mg dosage is supplied as either 5mg or 15mg capsules (six 5mg capsules or two 15mg capsules taken once daily by mouth) The 90mg dosage is supplied as 15mg capsules (six capsules, taken once daily by mouth)

T3D-959 3mg Experimental

Nine subjects will take 3mg by mouth once daily for two weeks, with or without food.

T3D-959 10mg Experimental

Nine subjects will take 10mg by mouth once daily for two weeks, with or without food.

T3D-959 30mg Experimental

Nine subjects will take 30mg by mouth once daily for two weeks, with or without food.

T3D-959 90mg Experimental

Nine subjects will take 90mg by mouth once daily for two weeks, with or without food.

Criteria

Inclusion Criteria:

Meets criteria for mild-to-moderate AD with Mini-Mental State Examination (MMSE) score of 14 through 26
Clinical Dementia Rating = 0.5 to 2.0
Modified Hachinski less than or equal to 4
A clinical diagnosis of AD per NINCDS-ADRDA criteria
Washout of psychoactive medication (other than anti-depressants): at least 4 weeks prior to baseline
Stability of all permitted medications for 4-12 weeks prior to baseline
Visual and auditory acuity adequate for neuropsychological testing
Home monitoring available for supervision of medications

Exclusion Criteria:

Unstable diabetes or insulin use
Unable to participate in FDG-PET scanning
Inability to undergo a clinical MRI of the brain
Diagnosis of significant neurological/psychiatric disease other than AD
History of moderate or severe congestive heart failure, NYHA class III or IV, within 12 months prior to baseline.
Previous cardiovascular event within the past 6 months prior to baseline
Subject is pregnant, or lactating.
ALT and/or AST levels that are twice the upper limit of normal; bilirubin levels that exceed 2 mg/dL; serum creatinine >1.5 mg/dL in men or > 1.4 mg/dL in women.
Current or history of severe or unstable disorder (medical or psychiatric) requiring treatment that may make the subject unlikely to complete the study.
Current use of fluvoxamine.
Current unstable use of warfarin.
Current use (within 30 days of baseline, visit 2) of certain highly protein-bound medications
Malignancy within the last 5 years (other than non-melanoma skin cancer, stable, non-progressive prostate cancer not requiring treatment or in situ cervical cancer).
Known history of HIV, hepatitis B, or hepatitis C.
Blood pressure greater than 160/100 mmHg.
Known or suspected intolerance or hypersensitivity to the study drugs, closely related compounds, or any of their stated ingredients.
History of alcohol, drug abuse or dependence (except nicotine dependence) within 2 years.
Investigational amyloid lowering therapies use within two months prior to baseline
Have participated in any other investigational study or received an investigational drug within 30 days or 5 half-lives (whichever is longer) prior to baseline
Any surgical or medical condition which may significantly alter the absorption of any drug substance
Resides in hospital or moderate to high dependency continuous care facility.
Non ambulatory, or wheelchair-bound
History of swallowing difficulties.
Evidence of clinically relevant pathology that in the investigator's opinion could interfere with the study results or put the subject's safety at risk.

Expanded Access Extension :

Subjects must continue to meet the main study inclusion/exclusion criteria to insure continued safety to continue on a 6 months study extension

Summary

T3D-959 3mg

T3D-959 10mg

T3D-959 30mg

T3D-959 90mg

All Events

Event Type Organ System Event Term T3D-959 3mg T3D-959 10mg T3D-959 30mg T3D-959 90mg

Change From Baseline (End of Treatment - Baseline) for FDG-PET Imaging With Whole Brain and White Matter as Reference Region

Changes in relative brain glucose metabolism (delta R CMRgl) were measured by FDG-PET. At each time point, a ratio of the PET reading in a pre-defined region of interest (sROI), known to be affected by AD, and in a reference region (RR) that is spared in AD, is determined. This ratio is defined as "sROI index" (spared region). A second RR, brain white matter (WM), was also used in this calculation: sROI index" (WM) value. delta sROI is defined as change in the sROI index values, over the treatment period. In this study we are looking for changes in delta sROI with increasing doses of T3D-959. Dose dependent changes in delta sROI (AD spared) are compared to those observed with the WM as the RR: delta sROI (WM). Dose related changes in delta sROI suggests T3D-959 is entering the brain and effecting glucose metabolism in a dose dependent fashion.

T3D-959 3mg

delta sROI (AD spared)

0.0015
ratio (Mean)
Standard Deviation: 0.02057

delta sROI (white matter)

0.0016
ratio (Mean)
Standard Deviation: 0.04008

T3D-959 10mg

delta sROI (AD spared)

0.0034
ratio (Mean)
Standard Deviation: 0.01671

delta sROI (white matter)

0.0053
ratio (Mean)
Standard Deviation: 0.02471

T3D-959 30mg

delta sROI (AD spared)

-0.0204
ratio (Mean)
Standard Deviation: 0.01952

delta sROI (white matter)

-0.02
ratio (Mean)
Standard Deviation: 0.01979

T3D-959 90mg

delta sROI (AD spared)

-0.0293
ratio (Mean)
Standard Deviation: 0.01744

delta sROI (white matter)

-0.0355
ratio (Mean)
Standard Deviation: 0.02303

The Effect of Treatment With T3D-959 on Changes in Resting State Blood Oxygen Level Dependent (BOLD) Signal in Functional Magnetic Resonance Imaging (fMRI) of the Brain Areas Associated With Cognitive Tasks.

Changes in BOLD fMRI parameters such as GoF (see Study Description) over the course of two weeks of treatment, were obtained in this study. BOLD fMRI has been used in cross sectional and longitudinal studies of Alzheimer's subjects, for instance in the Alzheimer's Disease Neuroimaging Initiative studies. However, no studies monitoring Default Mode Networks measured parameters such as GoF, in the context of an effective AD therapeutic, as a result it is difficult to interpret the observed small changes listed in BOLD fMRI parameters obtained in this trial. Instead the changes in the listed BOLD fMRI parameters (EOT - BL) are reported without interpretation. These values represent changes in fMRI connectivity patterns over time and are unitless.

T3D-959 3mg

ALFF_lIPL (ALFF from left inferior parietal lobule

-0.0635
unitless (Mean)
Standard Deviation: 0.09445

ALFF_lPCC_PreC (Amplitude of Low Frequency Fluctua

-0.0578
unitless (Mean)
Standard Deviation: 0.12314

ALFF_rIPL(ALFF from right IPL)

-0.0493
unitless (Mean)
Standard Deviation: 0.11288

ALFF_rPCC_PreC (ALFF from right PCC and precuneus)

-0.0752
unitless (Mean)
Standard Deviation: 0.09059

fALFF_lIPL (Ratio ALFF from left IPL)

-0.0296
unitless (Mean)
Standard Deviation: 0.03988

fALFF_lPCC_PreC (Ratio ALFF from left PCC & prec

0.0121
unitless (Mean)
Standard Deviation: 0.03246

fALFF_rIPL (Ratio ALFF from right IPL)

-0.0342
unitless (Mean)
Standard Deviation: 0.03912

fALFF_rPCC_PreC (Ratio ALFF from right PCC & pre

-0.008
unitless (Mean)
Standard Deviation: 0.04094

GlobEff_AAL(Global Efficiency from AAL Regions)

-0.0001
unitless (Mean)
Standard Deviation: 0.00664

GlobEff_DMN(Global Efficiency from DMN Regions)

0.0033
unitless (Mean)
Standard Deviation: 0.00734

GoF (Goodness of Fit)

0.0129
unitless (Mean)
Standard Deviation: 0.08528

Hippo-PreC Link (Hippocampus - Precuneus Link)

-0.1213
unitless (Mean)
Standard Deviation: 0.17904

ReHo_lIPL (Regional Homogeneity in left IPL)

-0.0694
unitless (Mean)
Standard Deviation: 0.15420

ReHo_lPCC_PreC (Regional Homogeneity in left PCC a

-0.0665
unitless (Mean)
Standard Deviation: 0.18502

ReHo_rIPL (Regional Homogeneity in right IPL)

-0.0694
unitless (Mean)
Standard Deviation: 0.15420

ReHo_rPCC_PreC (Regional Homogeneity in right PCC

-0.039
unitless (Mean)
Standard Deviation: 0.06406

T3D-959 10mg

ALFF_lIPL (ALFF from left inferior parietal lobule

0.0104
unitless (Mean)
Standard Deviation: 0.05763

ALFF_lPCC_PreC (Amplitude of Low Frequency Fluctua

-0.019
unitless (Mean)
Standard Deviation: 0.11983

ALFF_rIPL(ALFF from right IPL)

-0.0276
unitless (Mean)
Standard Deviation: 0.12720

ALFF_rPCC_PreC (ALFF from right PCC and precuneus)

-0.0127
unitless (Mean)
Standard Deviation: 0.12127

fALFF_lIPL (Ratio ALFF from left IPL)

0.0164
unitless (Mean)
Standard Deviation: 0.02269

fALFF_lPCC_PreC (Ratio ALFF from left PCC & prec

0.0068
unitless (Mean)
Standard Deviation: 0.03510

fALFF_rIPL (Ratio ALFF from right IPL)

0.0061
unitless (Mean)
Standard Deviation: 0.03286

fALFF_rPCC_PreC (Ratio ALFF from right PCC & pre

0.0084
unitless (Mean)
Standard Deviation: 0.04325

GlobEff_AAL(Global Efficiency from AAL Regions)

-0.0006
unitless (Mean)
Standard Deviation: 0.00972

GlobEff_DMN(Global Efficiency from DMN Regions)

-0.0036
unitless (Mean)
Standard Deviation: 0.00940

GoF (Goodness of Fit)

-0.012
unitless (Mean)
Standard Deviation: 0.0646

Hippo-PreC Link (Hippocampus - Precuneus Link)

0.132
unitless (Mean)
Standard Deviation: 0.22964

ReHo_lIPL (Regional Homogeneity in left IPL)

0.0147
unitless (Mean)
Standard Deviation: 0.14620

ReHo_lPCC_PreC (Regional Homogeneity in left PCC a

0.0237
unitless (Mean)
Standard Deviation: 0.14028

ReHo_rIPL (Regional Homogeneity in right IPL)

0.0147
unitless (Mean)
Standard Deviation: 0.14620

ReHo_rPCC_PreC (Regional Homogeneity in right PCC

-0.0663
unitless (Mean)
Standard Deviation: 0.22082

T3D-959 30mg

ALFF_lIPL (ALFF from left inferior parietal lobule

-0.0102
unitless (Mean)
Standard Deviation: 0.12016

ALFF_lPCC_PreC (Amplitude of Low Frequency Fluctua

-0.0505
unitless (Mean)
Standard Deviation: 0.07463

ALFF_rIPL(ALFF from right IPL)

0.0346
unitless (Mean)
Standard Deviation: 0.09654

ALFF_rPCC_PreC (ALFF from right PCC and precuneus)

-0.0097
unitless (Mean)
Standard Deviation: 0.08362

fALFF_lIPL (Ratio ALFF from left IPL)

-0.0107
unitless (Mean)
Standard Deviation: 0.06563

fALFF_lPCC_PreC (Ratio ALFF from left PCC & prec

-0.0132
unitless (Mean)
Standard Deviation: 0.03955

fALFF_rIPL (Ratio ALFF from right IPL)

0.0023
unitless (Mean)
Standard Deviation: 0.02771

fALFF_rPCC_PreC (Ratio ALFF from right PCC & pre

-0.0007
unitless (Mean)
Standard Deviation: 0.03913

GlobEff_AAL(Global Efficiency from AAL Regions)

-0.0031
unitless (Mean)
Standard Deviation: 0.01060

GlobEff_DMN(Global Efficiency from DMN Regions)

-0.0004
unitless (Mean)
Standard Deviation: 0.00926

GoF (Goodness of Fit)

0.0118
unitless (Mean)
Standard Deviation: 0.03375

Hippo-PreC Link (Hippocampus - Precuneus Link)

0.1196
unitless (Mean)
Standard Deviation: 0.23035

ReHo_lIPL (Regional Homogeneity in left IPL)

-0.0249
unitless (Mean)
Standard Deviation: 0.19178

ReHo_lPCC_PreC (Regional Homogeneity in left PCC a

-0.0369
unitless (Mean)
Standard Deviation: 0.09702

ReHo_rIPL (Regional Homogeneity in right IPL)

-0.0249
unitless (Mean)
Standard Deviation: 0.19178

ReHo_rPCC_PreC (Regional Homogeneity in right PCC

-0.0895
unitless (Mean)
Standard Deviation: 0.10541

T3D-959 90mg

ALFF_lIPL (ALFF from left inferior parietal lobule

-0.0401
unitless (Mean)
Standard Deviation: 0.05657

ALFF_lPCC_PreC (Amplitude of Low Frequency Fluctua

-0.0206
unitless (Mean)
Standard Deviation: 0.10508

ALFF_rIPL(ALFF from right IPL)

-0.1089
unitless (Mean)
Standard Deviation: 0.13935

ALFF_rPCC_PreC (ALFF from right PCC and precuneus)

-0.0325
unitless (Mean)
Standard Deviation: 0.11092

fALFF_lIPL (Ratio ALFF from left IPL)

-0.035
unitless (Mean)
Standard Deviation: 0.04570

fALFF_lPCC_PreC (Ratio ALFF from left PCC & prec

-0.0167
unitless (Mean)
Standard Deviation: 0.03284

fALFF_rIPL (Ratio ALFF from right IPL)

-0.0395
unitless (Mean)
Standard Deviation: 0.05327

fALFF_rPCC_PreC (Ratio ALFF from right PCC & pre

-0.0118
unitless (Mean)
Standard Deviation: 0.04236

GlobEff_AAL(Global Efficiency from AAL Regions)

-0.0005
unitless (Mean)
Standard Deviation: 0.00517

GlobEff_DMN(Global Efficiency from DMN Regions)

-0.0001
unitless (Mean)
Standard Deviation: 0.00397

GoF (Goodness of Fit)

0.0076
unitless (Mean)
Standard Deviation: 0.08755

Hippo-PreC Link (Hippocampus - Precuneus Link)

-0.0349
unitless (Mean)
Standard Deviation: 0.18496

ReHo_lIPL (Regional Homogeneity in left IPL)

-0.0062
unitless (Mean)
Standard Deviation: 0.06575

ReHo_lPCC_PreC (Regional Homogeneity in left PCC a

0.0194
unitless (Mean)
Standard Deviation: 0.20061

ReHo_rIPL (Regional Homogeneity in right IPL)

-0.0062
unitless (Mean)
Standard Deviation: 0.06575

ReHo_rPCC_PreC (Regional Homogeneity in right PCC

-0.0041
unitless (Mean)
Standard Deviation: 0.15836

Change From Baseline in the Score of the Digit Symbol Substitution Test

The digit symbol substitution test assesses attention, psychomotor speed, complex scanning, visual tracking, and immediate memory. This test consists of 4 rows each with 25 small blank squares; above each square is a number between 1 and 9. At the top is a 'key,' which pairs each number (1 through 9) with an unfamiliar symbol. The participant has 90 seconds to work as quickly as possible (left to right across the rows) to fill in each blank square with the appropriate symbol based on the number above the square. Results are presented as total number correct; therefore, lower numbers indicate greater impairment. Scores on the DSST range from 0-93.

T3D-959 3mg

Change from baseline at end of treatment (day 14)

4.429
score on a scale (Mean)
Standard Deviation: 9.1626

Change from baseline at follow-up (day 21)

5.375
score on a scale (Mean)
Standard Deviation: 8.5011

T3D-959 10mg

Change from baseline at end of treatment (day 14)

1.0
score on a scale (Mean)
Standard Deviation: 5.500

Change from baseline at follow-up (day 21)

3.111
score on a scale (Mean)
Standard Deviation: 5.0111

T3D-959 30mg

Change from baseline at end of treatment (day 14)

0.75
score on a scale (Mean)
Standard Deviation: 3.6154

Change from baseline at follow-up (day 21)

4.0
score on a scale (Mean)
Standard Deviation: 5.6789

T3D-959 90mg

Change from baseline at end of treatment (day 14)

1.125
score on a scale (Mean)
Standard Deviation: 4.5493

Change from baseline at follow-up (day 21)

6.625
score on a scale (Mean)
Standard Deviation: 11.4385

Change From Baseline in the Total Score of the 11-item Alzheimer's Disease Assessment Scale - Cognitive Subscale

The 11-item Alzheimer's Disease Assessment Scale (ADAS-Cog 11) is a psychometric instrument that evaluates memory, attention, reasoning, language, orientation, and praxis. The score can range between 0 and 70. A higher score indicates more cognitive impairment. A positive change in the score indicates cognitive worsening. The minimum severity score is 0 and the maximum severity score is 70.

T3D-959 3mg

Change from baseline at end of treatment (day 14)

-2.367
score on a scale (Mean)
Standard Deviation: 4.0417

Change from baseline at follow-up (day 21)

-3.409
score on a scale (Mean)
Standard Deviation: 3.5168

T3D-959 10mg

Change from baseline at end of treatment (day 14)

-0.552
score on a scale (Mean)
Standard Deviation: 3.1467

Change from baseline at follow-up (day 21)

-0.624
score on a scale (Mean)
Standard Deviation: 3.8134

T3D-959 30mg

Change from baseline at end of treatment (day 14)

-2.709
score on a scale (Mean)
Standard Deviation: 3.9801

Change from baseline at follow-up (day 21)

-4.119
score on a scale (Mean)
Standard Deviation: 3.8163

T3D-959 90mg

Change from baseline at end of treatment (day 14)

1.97
score on a scale (Mean)
Standard Deviation: 5.2191

Change from baseline at follow-up (day 21)

2.795
score on a scale (Mean)
Standard Deviation: 4.1567

Safety and Tolerability of Treatment With T3D-959 Over a 2-week Period in Subjects With Mild-to-moderate AD. New

Number of participants with treatment related adverse events (AEs) as assessed by analysis of adverse events, including symptoms, and abnormal findings on physical and neurological examinations, and standard labs.

T3D-959 3mg

Subjects with at Least One Drug-related AE

Subjects with at Least One mild AE

2.0
participants

Subjects with at Least One moderate AE

1.0
participants

Subjects with at Least One Serious Adverse Events

Subjects with at Least One serious AE

T3D-959 10mg

Subjects with at Least One Drug-related AE

Subjects with at Least One mild AE

Subjects with at Least One moderate AE

Subjects with at Least One Serious Adverse Events

Subjects with at Least One serious AE

T3D-959 30mg

Subjects with at Least One Drug-related AE

1.0
participants

Subjects with at Least One mild AE

1.0
participants

Subjects with at Least One moderate AE

2.0
participants

Subjects with at Least One Serious Adverse Events

Subjects with at Least One serious AE

T3D-959 90mg

Subjects with at Least One Drug-related AE

Subjects with at Least One mild AE

Subjects with at Least One moderate AE

Subjects with at Least One Serious Adverse Events

Subjects with at Least One serious AE

Total

36
Participants

Age, Customized

ApoE4 genotype

Sex: Female, Male

Overall Study

T3D-959 3mg

T3D-959 10mg

T3D-959 30mg

T3D-959 90mg