SBIR-STTR Award

Multiple Ascending Dose Clinical Trial of PTI-125, a Novel AD Therapeutic Candidate
Award last edited on: 5/14/2020

Sponsored Program
SBIR
Awarding Agency
NIH : NIA
Total Award Amount
$3,906,146
Award Phase
2
Solicitation Topic Code
-----

Principal Investigator
Lindsay H Burns

Company Information

Cassava Sciences Inc (AKA: Pain Therapeutics Inc)

6801 N Capital Of Texas Hwy Bldg 1 Suite 300
Austin, TX 78731
   (512) 501-2444
   cwaarich@paintrials.com
   www.paintrials.com
Location: Single
Congr. District: 37
County: Travis

Phase I

Contract Number: 1R44AG060878-01
Start Date: 9/1/2018    Completed: 10/31/2018
Phase I year
2018
Phase I Amount
$260,585
PTI-125 is a novel small molecule Alzheimer's disease (AD) therapeutic candidate with a novel target and mechanism of action. PTI-125 binds and reverses an altered conformation of the scaffolding protein filamin A (FLNA) to prevent A?42's tight binding to and toxic signaling via the ?7-nicotinic acetylcholine receptor (?7nAChR) as well as A?42's aberrant activation of toll-like receptor 4 (TLR4). Hence, by restoring FLNA's native shape and blocking these two toxic cascades, PTI-125 reduces both tau hyperphosphorylation and neuroinflammation. Downstream effects include reduced neurofibrillary lesions and amyloid deposits, suggesting disease modification, and improved synaptic plasticity and function of ?7nAChR, NMDAR and insulin receptors, suggesting symptomatic improvement. We will pursue a label claim of symptomatic improvement instead of the more difficult claim of disease modification and will therefore conduct clinical studies in mild-to-moderate AD. Under a US IND, the first-in-human clinical trial showed no drug- related adverse effects (AEs) and dose proportional pharmacokinetics (PK) of the oral solution. In this fast-track proposal, we will first conduct CYP interaction studies to determine potential drug-drug interactions of PTI-125. CYP interaction studies were requested by FDA in pre-IND guidance. This information will be incorporated into the protocol for a 1-month multi-dose clinical trial in mild-to-moderate AD patients for safety and PK in Phase II of this application. To demonstrate target engagement, we will use our plasma-based companion diagnostic/biomarker as well as an earlier lymphocyte- based version that tracked treatment effects in mice. This 1-month study will also include the neuroinflammation biomarker YKL40 and efficacy endpoints of cognition, and irritability/agitation. The clinical trial proposed here will guide subsequent Phase 2 clinical trials and make PTI-125 more attractive to potential commercialization partners.

Public Health Relevance Statement:
Project Narrative PTI-125 is a novel compound has been shown to alleviate multiple pathological features of AD in mouse models and in postmortem AD brain tissue, including receptor dysfunctions, inflammation, impaired synaptic plasticity, and the hallmark plaques and tangles. These multiple therapeutic benefits suggest PTI-125 may improve cognitive function as well as slow the course of the disease. Following CYP interaction studies, we propose to conduct a 1-month multi-dose PK and safety clinical trial in mild-to-moderate AD patients that will also collect plasma and CSF biomarkers as well as efficacy endpoints for cognition and irritability.

NIH Spending Category:
Acquired Cognitive Impairment; Aging; Alzheimer's Disease; Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD); Brain Disorders; Clinical Research; Clinical Trials and Supportive Activities; Dementia; Neurodegenerative; Neurosciences; Precision Medicine

Project Terms:
Adverse effects; Affinity; Aftercare; Age; Agitation; alpha-bungarotoxin receptor; Alzheimer's Disease; Amyloid; Amyloid beta-Protein; Autopsy; base; Binding; Bioavailable; Biological Assay; Biological Markers; Blood; Body Surface Area; Brain; Brain Diseases; brain tissue; Canis familiaris; CD14 gene; Chemistry; Clinical; Clinical Research; Clinical Trials; Cognition; Cognitive; cognitive function; commercialization; companion diagnostics; Companions; Cytochrome P450; cytokine; Data; Deposition; diagnostic biomarker; Disease; Disease Progression; Dose; Drug Interactions; Drug Kinetics; Electrocardiogram; enzyme activity; Equipment and supply inventories; exposed human population; filamin; first-in-human; Functional disorder; Grapefruit; healthy volunteer; Hematology; Hepatic; Hepatocyte; Human; Human Resources; hyperphosphorylated tau; Impairment; improved; indexing; Inflammation; Inflammatory; Insulin Receptor; Label; Lymphocyte; Measurement; Measures; Mediating; Microsomes; Modification; Molecular Conformation; mouse model; mRNA Expression; Mus; NADP; Neurofibrillary Tangles; neuroinflammation; neuropsychiatry; No-Observed-Adverse-Effect Level; novel; Oral; Pathologic; patient safety; Patients; Pharmaceutical Preparations; Phase; Phase II Clinical Trials; Phosphotransferases; Placebos; Plasma; prevent; Protocols documentation; Rattus; receptor; Safety; safety study; Scaffolding Protein; Shapes; Signal Transduction; small molecule; small molecule therapeutics; symptomatic improvement; synaptic function; Synaptic plasticity; tau Proteins; Testing; Therapeutic; therapeutic candidate; Time; TLR4 gene; Toxic effect; Toxicology; Transgenic Mice; treatment effect; Urinalysis; Work

Phase II

Contract Number: 4R44AG060878-02
Start Date: 00/00/00    Completed: 00/00/00
Phase II year
2019
(last award dollars: 2020)
Phase II Amount
$3,645,561

PTI-125 is a novel small molecule Alzheimer's disease (AD) therapeutic candidate with a novel target and mechanism of action. PTI-125 binds and reverses an altered conformation of the scaffolding protein filamin A (FLNA) to prevent A?42's tight binding to and toxic signaling via the ?7-nicotinic acetylcholine receptor (?7nAChR) as well as A?42's aberrant activation of toll-like receptor 4 (TLR4). Hence, by restoring FLNA's native shape and blocking these two toxic cascades, PTI-125 reduces both tau hyperphosphorylation and neuroinflammation. Downstream effects include reduced neurofibrillary lesions and amyloid deposits, suggesting disease modification, and improved synaptic plasticity and function of ?7nAChR, NMDAR and insulin receptors, suggesting symptomatic improvement. We will pursue a label claim of symptomatic improvement instead of the more difficult claim of disease modification and will therefore conduct clinical studies in mild-to-moderate AD. Under a US IND, the first-in-human clinical trial showed no drug- related adverse effects (AEs) and dose proportional pharmacokinetics (PK) of the oral solution. In this fast-track proposal, we will first conduct CYP interaction studies to determine potential drug-drug interactions of PTI-125. CYP interaction studies were requested by FDA in pre-IND guidance. This information will be incorporated into the protocol for a 1-month multi-dose clinical trial in mild-to-moderate AD patients for safety and PK in Phase II of this application. To demonstrate target engagement, we will use our plasma-based companion diagnostic/biomarker as well as an earlier lymphocyte- based version that tracked treatment effects in mice. This 1-month study will also include the neuroinflammation biomarker YKL40 and efficacy endpoints of cognition, and irritability/agitation. The clinical trial proposed here will guide subsequent Phase 2 clinical trials and make PTI-125 more attractive to potential commercialization partners.

Public Health Relevance Statement:
Project Narrative PTI-125 is a novel compound has been shown to alleviate multiple pathological features of AD in mouse models and in postmortem AD brain tissue, including receptor dysfunctions, inflammation, impaired synaptic plasticity, and the hallmark plaques and tangles. These multiple therapeutic benefits suggest PTI-125 may improve cognitive function as well as slow the course of the disease. Following CYP interaction studies, we propose to conduct a 1-month multi-dose PK and safety clinical trial in mild-to-moderate AD patients that will also collect plasma and CSF biomarkers as well as efficacy endpoints for cognition and irritability.

NIH Spending Category:
Acquired Cognitive Impairment; Aging; Alzheimer's Disease; Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD); Brain Disorders; Clinical Research; Clinical Trials and Supportive Activities; Dementia; Neurodegenerative; Neurosciences; Precision Medicine

Project Terms:
Adverse effects; Affinity; Aftercare; Age; Agitation; alpha-bungarotoxin receptor; Alzheimer's Disease; Amyloid; Amyloid beta-Protein; Autopsy; base; Binding; Bioavailable; Biological Assay; Biological Markers; Blood; Body Surface Area; Brain; Brain Diseases; brain tissue; Canis familiaris; CD14 gene; Chemistry; Clinical; Clinical Research; Clinical Trials; Cognition; Cognitive; cognitive function; commercialization; companion diagnostics; Companions; Cryopreservation; Cytochrome P450; cytokine; Data; Deposition; diagnostic biomarker; Disease; Disease Progression; Dose; Drug Interactions; Drug Kinetics; Electrocardiogram; enzyme activity; Equipment and supply inventories; exposed human population; filamin; first-in-human; Functional disorder; Grapefruit; healthy volunteer; Hematology; Hepatic; Hepatocyte; Human; Human Resources; hyperphosphorylated tau; Impairment; improved; indexing; Inflammation; Inflammatory; Insulin Receptor; Label; Lymphocyte; Measurement; Measures; Mediating; Microsomes; Modification; Molecular Conformation; mouse model; mRNA Expression; Mus; NADP; Neurofibrillary Tangles; neuroinflammation; neuropsychiatry; No-Observed-Adverse-Effect Level; novel; Oral; Pathologic; patient safety; Patients; Pharmaceutical Preparations; Phase; Phase II Clinical Trials; Phosphotransferases; Placebos; Plasma; prevent; Protocols documentation; Rattus; receptor; Safety; safety study; Scaffolding Protein; Shapes; Signal Transduction; small molecule; small molecule therapeutics; symptomatic improvement; synaptic function; Synaptic plasticity; tau Proteins; Testing; Therapeutic; therapeutic candidate; Time; TLR4 gene; Toxic effect; Toxicology; Transgenic Mice; treatment effect; Urinalysis; Work