Research Models

SOD1 (G37R)

Synonyms: line 29, SOD1 (G37R) (hybrid background), G37R(29) SOD1, SOD1 G37R

Tools

Back to the Top

Species: Mouse
Genes: SOD1
Modification: SOD1: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis
Strain Name: N/A

Summary

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

  • Cortical Neuron Loss
  • Cytoplasmic Inclusions

No Data

Cortical Neuron Loss

Upper motor neuron loss was not observed, although vacuolization occurred in brainstem neurons.

Lower Motor Neuron Loss

Motor neurons in the spinal cord and brainstem degenerated with overt neuronal loss in the ventral horn in some regions of the spinal cord by 19 weeks. The degenerative process involved extensive vacuolization.

Cytoplasmic Inclusions

Not observed.

Gliosis

Astrogliosis occurs in the spinal cord by 11 weeks of age, becoming more severe with age.

NMJ Abnormalities

Denervated endplates have been observed.

Muscle Atrophy

Loss of motor axons, denervated endplates, atrophy of muscle fibers, and fiber type grouping observed by end-stage.

Motor Impairment

Motor impairment at 4-6 months, beginning with reduced spontaneous movement, then tremors, limb weakness, and poor grooming. Eventual paralysis of the hindlimbs.

Body Weight

Loss of body weight is observed.

Premature Death

Mice survive about 6 to 8 months.

Last Updated: 06 Mar 2018

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

CT1812

Tools

Back to the Top

Overview

Name: CT1812
Synonyms: Elayta
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: Cognition Therapeutics Inc.

Background

CT1812 is a small-molecule antagonist of the sigma2 receptor. The rationale behind this therapeutic approach is that ligands for the sigma2 receptor complex are negative allosteric regulators that reduce the affinity of oligomeric Aβ for neuronal receptors, and thus interfere with Aβ-induced synaptic toxicity.

CT1812 grew out of screening programs at Cognition Therapeutics. Company scientists have reported that compounds in this series not only block binding of a range of different Aβ species to neuronal receptors but also displace it when applied after Aβ has bound (Dec 2014 conference news). Subsequently, company scientists reported the compounds also inhibit the ability of α-synuclein oligomers to induce lysosomal trafficking defects in rat neurons and glia (Limegrover et al., 2021).

CT1812 was chemically optimized from a series of small molecules selected for their ability to reverse the synaptotoxicity of soluble Aβ oligomers on cultured neurons (Rishton et al., 2021). Similar compounds in the series have been reported to enter the brain, occupy up to 80 percent of sigma2 receptors, and restore behavioral deficits in APP transgenic mice (Izzo et al., 2014Izzo et al., 2014). In preclinical work, CT1812 facilitated brain clearance of Aβ oligomers and improved cognitive behaviors in transgenic mouse models of Alzheimer’s disease. The compound also displaced Aβ oligomers from human postmortem AD brain tissue (Izzo et al., 2021).

The identification of the gene encoding the sigma2 protein has allowed more detailed mechanistic studies on the receptor and the actions of CT1812 (Alon et al., 2017). It was reported that a complex of the sigma 2 protein, progesterone receptor membrane component 1, and the low-density lipoprotein receptor controls the internalization of Aβ monomers and oligomers in synapses (Riad et al., 2020). Further studies found evidence for increased synaptic levels of sigma2, also known as TMEM97, in Alzheimer’s disease (Hesse et al., 2019). FRET imaging suggests a close synaptic interaction between Aβ oligomers and TMEM97 in Alzheimer’s brain tissue. In mouse and human models, CT1812 inhibited this interaction, and also caused changes in the expression of genes related to synaptic function (Colom-Cadena et al., 2024).

Findings

From September 2015 to May 2016, Cognition Therapeutics ran a Phase 1 trial in 80 healthy volunteers aged 18 to 75 in Melbourne, Australia; target enrollment was originally listed as 114. Single-ascending-dose administration was followed by multiple ascending doses given once daily for two weeks. The dose range in this trial spanned 10 to 650 mg; if this would not generate data to set a maximum tolerated dose, doses up to 1,350 mg were to be tried. Outcome measures included safety, tolerability, plasma pharmacokinetics, and CSF CT1812 concentration. At the 2016 and 2017 AAIC conferences, company scientists reported that single doses up to 1,120 mg were given, as were multiple doses of up to 840 mg in young and up to 560 mg in elderly volunteers. The drug was reported to be well-tolerated, with suitable pharmacokinetics, sufficient brain penetrance and target exposure, and minimal drug-drug interactions affecting cytochrome P450 activity (Catalano et al., 2016). Results have been published (Grundman et al., 2019). Another Phase 1 trial in 2016 assessed drug-drug interactions in 16 healthy volunteers.

From September 2016 to August 2017, Cognition Therapeutics ran a Phase 1/2 trial at four sites in Australia, enrolling 19 participants with mild to moderate Alzheimer's disease supported by a recent MRI. It compared a four-week course of 90, 280, or 560 mg of CT1812 to placebo, taken once daily, on safety and tolerability parameters. At the subsequent CTAD conference, Elayta was reported to have been generally safe and well-tolerated, though there were four cases of lymphocytopenia. Exploratory measures such as ADAS-Cog14, verbal or category fluency tests recorded no difference between groups, but exploratory biomarker analyses yielded possible signals of synapse protection (Dec 2017 conference news). Later, investigators reported that CT1812 increased Aβ oligomers in CSF, suggesting CT1812 promoted oligomer clearance from the brain (2019 AAIC meeting abstract). According to published data, the Aβ oligomer concentration in CSF rose after treatment, while the concentration of synaptic proteins neurogranin and synaptotagmin fell, compared to placebo. CSF Aβ40 or 42 monomers, neurofilament light chain, SNAP-25, total Tau, and p-Tau181 were unchanged (Izzo et al., 2021).

In April 2018, a Phase 1/2 study started enrolling 23 people whose mild to moderate AD was confirmed by amyloid PET or CSF testing. Conducted at Yale University School of Medicine and dubbed COG0105 or SPARC, this trial compared a six-month course of 100 or 300 mg of Elayta to placebo. A six-month, double-blind extension was later added. The primary outcome was originally listed as cognition as assessed by the Alzheimer's Disease Clinical Study Activities of Daily Living (ADCS-ADL), but was changed in September 2018 to safety. The trial used the investigational PET tracer UCB-J, which binds to the synaptic vesicle glycoprotein 2A, in an attempt to monitor synapse density before and after treatment (see company press release; Jul 2016 news). Midway through the study, synaptic density was added as co-primary. The trial finished in October 2020 after enrolling 43 patients, and results are posted on clinicaltrials.gov. At the 2022 AAIC, the company disclosed that treatment had not altered synaptic density, measured by SV2a PET, compared to baseline. CSF proteomics analysis revealed a reduction in the inflammatory marker YKL-40. Other CSF proteins related to synaptic biology and amyloid were reportedly normalized after treatment (company presentation). According to published results, treatment caused no change in FDG-PET, clinical cognitive scales, or CSF biomarkers (Van Dyck et al., 2024). Volumetric MRI revealed a trend toward tissue preservation with treatment.

In May 2018, a Phase 1b target engagement study at the University of Pennsylvania began enrolling 18 people whose mild to moderate AD was to be confirmed by amyloid PET. Called COG0104 or SNAP, the study was to compare single injections of 90, 280, or 560 mg of Elayta to placebo for their ability to displace Aβ oligomers and clear them into the CSF. This was assessed by an Aβ oligomer assay on CSF samples collected hourly for 24 hours after treatment. This study ended in February 2019, with only three participants enrolled. According to published results, the investigators had difficulty recruiting because of the study's requirement for a 28-hour spinal catherization and the lack of an optional open-label extension (LaBarbera et al., 2023). Of the three participants, two received 560 mg CT1812, one placebo. CSF Aβ oligomers increased by 2.5- and fivefold over baseline in the treated patients, and did not change in the one who received placebo. Concentrations of monomeric Aβ stayed the same in all three. One patient who had higher blood levels of CT1812 also had higher CSF Aβ oligomer concentrations.

A similar pilot trial in Sweden was to measure CSF Aβ oligomers after a single 100 mg dose in 16 people with mild to moderate AD, followed by six months of open-label daily dosing. The trial was to assess drug effects on CSF biomarkers of Aβ, tau, and synaptic function after six months. In November 2020, this trial was stopped early.

In October 2018, a Phase 2 multicenter safety study called SHINE or COG0201 began enrolling 24 people with mild to moderate AD as confirmed by amyloid PET for a six-month course of 100 or 300 mg of CT1812, or placebo. The primary outcome is the number of adverse events. The company later increased the study size to 144, and added cognitive endpoints. The trial, at 36 sites in the U.S., Europe, and Australia, finished enrollment at 153 participants in November 2023 (press release). At the 2020 AAIC, the company reported positive trends for cognitive endpoints in the first 24 people who completed six months of treatment (Aug 2020 conference news). A proteomics analysis of CSF from 18 participants revealed treatment-related changes in proteins linked to synapses, lipoprotein and Aβ biology, and neuroinflammation (Lizama et al., 2024). The trial was completed in May 2024, and results presented on July 29 at AAIC (press release). The drug met its safety endpoint, with no discontinuations due to side effects in the 100 mg dose. Elevations in liver enzymes were detected in nine of the 42 patients on the 300 mg dose, and reversed when drug was stopped. On cognitive endpoints, CT1812 treatment slowed decline by about one point on the ADAS-Cog11 compared to placebo; the difference was not statistically significant. At the 300 mg dose, CSF NfL and Aβ42 did decline. Other markers of amyloid and tau pathology, inflammation, and synaptic function were unchanged.

In July 2020, a pilot study started in Amsterdam to assess synaptic effects of CT1812 using quantitative electroencephalography. Sixteen participants with mild to moderate Alzheimer’s disease receive 300 mg Elayta and placebo for 29 days each, in a crossover design. Outcomes include EEG, cognitive and functional measures, CSF biomarkers, and safety. The study was completed in April 2023 and results were presented at the October 2023 CTAD conference. All prespecified EEG parameters trended toward improvement; significant increases in theta power and functional connectivity suggested the drug may be able to enhance synaptic activity (Vijverberg et al., 2024).

In January 2022, the company conducted an eight-patient study of the absorption, metabolism, and secretion of a single oral dose of [14C]-CT1812; results are posted at clinicaltrials.gov. In February to June, they conducted a pharmacokinetic study in 35 older healthy volunteers, evaluating 150 mg once or twice a day, and 300 mg once a day.

In June 2022, a Phase 2 trial called SHIMMER began assessing CT1812 in 120 people with mild to moderate dementia with Lewy bodies. At 37 centers in the U.S., this six-month study is comparing 100 or 300 mg of CT1812 to placebo on primary outcomes of safety and tolerability. Secondary outcomes include measures of cognition, sleepiness, clinical change, daily function, movement, and neuropsychiatric symptoms. The study completed enrollment with 130 patients, and is planned to finish in November 2024.

In September 2022, the company registered a Phase 2 trial called START. It will compare 18 months of 100 or 300 mg CT1812 to placebo in 540 people with mild cognitive impairment or mild dementia due to Alzheimer’s. The primary outcome is change in CDR-SB, with planned secondaries of the ADAS-Cog13, ADCS-Activities of Daily Living, CSF biomarkers of Aβ, tau, neurofilament light chain, neurogranin, and synaptotagmin, as well as volumetric MRI. The Alzheimer’s Clinical Trial Consortium is running the trial, with funding from the National Institute on Aging (press release). After awaiting FDA clearance, the trial began in June 2023, and will run until spring 2027. According to a presentation at the October 2023 CTAD conference, inclusion criteria were modified to permit enrollment of people who are taking the since-approved anti-amyloid antibody lecanemab; however, the trial is not designed to be a concurrent or combination trial.

In June 2023, the company began a Phase 2 study testing CT1812 for age-related macular degeneration (press release).

For all trials of this compound, see clinicaltrials.gov and EU Clinical Trials Register.

Clinical Trial Timeline

  • Phase 1/2
  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035
Cognition Therapeutics Inc. NCT02907567
N=19
Cognition Therapeutics Inc. NCT03493282
N=21
Cognition Therapeutics Inc. NCT03507790
N=24

Last Updated: 15 Aug 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

TDP-43 (WT) (Kumar-Singh)

Synonyms: WT-TAR4/4, WT-TAR4

Tools

Back to the Top

Species: Mouse
Genes: TARDBP
Modification: TARDBP: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
Strain Name: B6;SJL-Tg(Thy1-TARDBP)4Singh/J

Summary

This mouse model of ALS overexpresses wild-type human TARDBP (TDP-43) in postnatal neurons. Mice in this line, called TAR4, develop ALS-related phenotypes, including severe motor impairment, loss of upper and lower motor neurons, and premature death. Homozygous mice (referred to as TAR4/4) become symptomatic very early and do not survive beyond one month of age. Hemizygous mice (referred to as TAR4) exhibit much milder phenotypes (Wils et al., 2010).

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

No Data

  • NMJ Abnormalities
  • Muscle Atrophy

Cortical Neuron Loss

In homozygous mice, quantitative loss of neurons occurs in the motor cortex compared with non-Tg littermates. Both superficial and deep cortical layers of the anterior cortex are affected.

Lower Motor Neuron Loss

By day 18, homozygous mice exhibited about 25 percent loss of motor neurons in the lumbar spinal cord compared with non-Tg littermates.

Cytoplasmic Inclusions

Homozygous mice developed cytoplasmic inclusions in the brain and spinal cord, many of which were ubiquitin-positive. A minority of inclusions co-labeled with TDP-43. Ultrastructural analysis revealed ubiquitin–negative cytoplasmic inclusions in anterior horn neurons to be abnormal accumulations of mitochondria.

Gliosis

Astrogliosis and microgliois especially in cortical layer V of the anterior cortex, including motor and somatosensory cortex, and in the spinal cord.

NMJ Abnormalities

No data.

Muscle Atrophy

No data.

Motor Impairment

Homozygous mice exhibit an abnormal clasping reflex by postnatal day 14. Other early motor deficits include a shortened stride, a wide stance, and frequent stumbling. By day 18, reduced performance on the Rotarod. Complete paralysis occurs ~10 days after onset.

Body Weight

Size and weight of homozygous mice lag behind hemizygotes and non-Tg littermates.

Premature Death

Homozygous mice survive an average of just 24 days. In contrast, hemizygous mice survive to advanced age, although they die more prematurely than non-Tg mice, after 22 to 24 months.

Last Updated: 03 Apr 2024

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Young Plasma

Tools

Back to the Top

Overview

Name: Young Plasma
Therapy Type: Other
Target Type: Inflammation (timeline), Other (timeline), Unknown
Condition(s): Progressive Supranuclear Palsy, Alzheimer's Disease
U.S. FDA Status: Progressive Supranuclear Palsy (Inactive), Alzheimer's Disease (Inactive)
Company: Alkahest, Inc., Grifols Biologicals Inc.

Background

The rationale behind this treatment approach is that systemic factors present in young blood cross into the brain, where they counteract aging processes and neurodegeneration by modulating neuroinflammation, neurogenesis, and cognitive function. In essence, this approach attempts to rejuvenate the aging brain with infusions of plasma from young adults. Initial clinical trials grew, in part, out of parabiosis studies, in which old mice connected via their circulatory systems to young mice showed improvements on a wide range of outcome measures related to aging and neurodegeneration (May 2014 conference newsMar 2013 conference newsNov 2009 conference news).

On February 2019, responding to reports of commercial delivery of young plasma as a purported treatment for various conditions including memory loss, the FDA issued a statement warning the public of the potential risks of this practice. GRF6019 is a plasma fraction from which clotting factors and immunoglobulins have been removed to reduce those risks.

Findings

From September 2014 to February 2017, a double-blinded Phase 1 study at Stanford University enrolled 18 people with mild to moderate Alzheimer's disease into a 12-week trial. It compared a four-week treatment regimen of once-weekly injections of one unit of plasma from men aged 30 or under to placebo. Treatment and placebo groups switched off after a four-week washout. Safety was the primary outcome; other outcomes included cognitive, functional, and neuropsychiatric AD scales, as well as functional connectivity in the default mode network as per resting state functional MRI. At the 2017 CTAD meeting, the treatment was reported to have been safe and to have yielded a signal on functional/daily activities tests. However, two participants dropped out and the study switched to open-label midway through (Dec 2017 conference news, and Sha et al., 2019). For details, see clinicaltrials.gov.  

In May 2015, a Phase 1 study at the University of California, San Francisco, enrolled six people with progressive supranuclear palsy into an open-label study of transfusion of plasma from blood donors 30 or younger. This study infuses four units of fresh frozen plasma once a month for six months. Beyond the primary outcome of safety, this trial lists 13 additional outcome measures ranging from change in motor, cognitive, functional, and behavioral measures to CSF, MRI, saccadic eye movement, and sleep measures. See clinicaltrials.gov. The trial was completed in December 2019, and results were published. The infusions were safe but did not change exploratory endpoints (VandeVrede et al., 2020).

From 2016-2019, a Phase 1 study of young plasma in 15 people with Parkinson’s disease was carried out at Stanford University. Infusions were safe and were reported to improve exploratory endpoints of verbal fluency (Parker et al., 2020).

In 2018, a second-generation approach to young plasma entered clinical trials, see GRF6019.

Last Updated: 14 Jan 2021

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

TDP-43 (A315T) (line 23)

Tools

Back to the Top

Species: Mouse
Genes: TARDBP
Modification: TARDBP: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
Strain Name: STOCK Tg(Prnp-TARDBP*A315T)23Jlel/J

Summary

This mouse model of ALS was among the first transgenics expressing mutant TDP-43 to be characterized (Stallings et al., 2010). In this model the mouse prion protein (Prp) promoter drives TDP-43 expression in the central nervous system, including in the brain and spinal cord, with lower expression elsewhere (i.e., skeletal muscle). Hemizygous mice develop progressive motor impairment, ultimately leading to paralysis and premature death at around 2.5 months of age.

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

  • Cortical Neuron Loss
  • Lower Motor Neuron Loss

No Data

  • NMJ Abnormalities

Cortical Neuron Loss

Not observed.

Lower Motor Neuron Loss

Not observed.

Cytoplasmic Inclusions

Ubiquitin-positive cytoplasmic inclusions in neurons of the ventral horn and brainstem. Cytoplasmic aggregates of TDP-43 are largely absent, although rare phospho-TDP-43 inclusions were observed, especially at end-stage.

Gliosis

Mice exhibiting muscle weakness had astrocytosis in the ventral horn of the spinal cord.

NMJ Abnormalities

Unknown.

Muscle Atrophy

Atrophy of muscle fibers in the quadriceps muscle of weak mice observed by day 44.

Motor Impairment

Progressive motor impairment, characterized by weakness, a decline in grip strength, and reduction in stride length. Weakness was usually more pronounced in the hindlimbs.

Body Weight

Progressive weight loss.

Premature Death

Line 23 mice survived about 2.5 months, mean survival 75 days. It was not reported whether this survival analysis includes males, females or both. Colony at Jackson Labs has longer mean survival.

Last Updated: 06 Mar 2018

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

TARDBP (A315T) (hybrid)

Synonyms: Baloh’s TDP-43, C57BL/6-CBA TDP-43 A315T

Tools

Back to the Top

Species: Mouse
Genes: TARDBP
Modification: TARDBP: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis
Strain Name: N/A

Summary

In 2009, Robert Baloh and colleagues reported a transgenic mouse model of ALS based on the overexpression of mutant TARDBP (TDP-43) (Wegorzewska et al., 2009). These mice developed several features of ALS, including motor impairment, degeneration of cortical and spinal motor neurons, cytoplasmic aggregates of ubiquitinated proteins, and premature death. Notably absent was cytoplasmic deposition of TDP-43 protein.

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

No Data

  • NMJ Abnormalities

Cortical Neuron Loss

By end-stage, neuronal numbers in layer 5 of the motor cortex are decreased with about 50 percent loss of corticospinal tract axons.

Lower Motor Neuron Loss

By end-stage, ~20% loss of motor neurons in the L3-L5 region of the spinal cord.

Cytoplasmic Inclusions

By end-stage, cytoplasmic inclusions of ubiquitinated proteins in layer 5 neurons of motor, sensory, and cingulate cortex. Ubiquitin aggregates in ventral horn neurons. TDP-43 inclusions were rare.

Gliosis

By end-stage, selective increase in GFAP immunoreactivity in cortical layer 5.

NMJ Abnormalities

Unknown.

Muscle Atrophy

By end-stage, atrophic muscle fibers were observed.

Motor Impairment

Gait abnormalities around three months of age, developing into a characteristic “swimming gait” by four to five months.

Body Weight

Weight was comparable to non-Tg mice at birth. By 4.5 months transgenic mice began to lose weight.

Premature Death

Survival for about 5 months (154 ± 19 days) before dying spontaneously or being euthanized. It was not reported if this analysis includes males, females, or both.

Last Updated: 04 Jun 2019

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

T3D-959

Tools

Back to the Top

Overview

Name: T3D-959
Synonyms: DB959
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: T3D Therapeutics, Inc.

Background

T3D-959 is a dual agonist of the peroxisome proliferator activated nuclear receptor delta/gamma, aka PPARδ/γ. It is being developed as an oral therapy for Alzheimer’s disease. In 2013, T3D Therapeutics acquired this compound from DARA BioSciences. In 2010 and 2011, DARA BioSciences had tested DB959 in Phase 1 trials for dyslipidemia and Type 2 diabetes, but subsequently shifted its focus to oncology care products.

The PPAR family of proteins helps regulate blood sugar and triglyceride levels. PPAR activation affects these measures by boosting insulin sensitivity, and this approach has become a mainstay for correcting insulin resistance in diabetes therapy. The rationale for evaluating PPAR agonists in Alzheimer's is based on the hypothesis that sporadic AD is fundamentally an age-related metabolic disease, sometimes called Type 3 diabetes (Sep 2006 conference newsde la Monte and Tong, 2014). Unlike other PPAR-targeted diabetes drugs that have been evaluated in Alzheimer's previously, T3D-959 is not a thiazolidinedione.

The peer-reviewed literature contains three preclinical research reports on T3D-959, by scientists affiliated with T3D Therapeutics. Two report treatment benefits on spatial navigation, memory, and motor performance in rats previously injected with streptozotocin, a pancreatic cancer chemotherapy that poisons insulin-producing beta cells and is used to model diabetes in rodents (Tong et al., 2016Tong et al., 2016). Another paper reports that T3D-0959 normalized streptozotocin-induced changes in the biomarkers pTau, AβPP, Aβ42, ubiquitin, SNAP-25, synaptophysin, IGF-1 receptor (R), IRS-1, Akt, p70S6K, mTOR, and S9-GSK-3β, also in rats (de la Monte et al., 2017).

Findings

In July 2015, a Phase 1/2 study enrolled 36 people with mild to moderate Alzheimer's disease to compare a two-week course of either 3, 10, 30, or 90 mg of T3D-959, taken once daily. This trial did not have a placebo group. It was a biomarker proof-of-mechanism study, in that the primary outcomes were change from baseline of cerebral glucose metabolism as measured by FDG-PET, and change from baseline of functional connectivity between the precuneus/posterior cingulate cortex and hippocampus as measured by resting-state fMRI. The Digit Symbol Substitution Test, ApoE subgroup analysis, blood lipid metabolomics, and ADAS-Cog 11 constituted secondary outcomes. Conducted in Florida and South Carolina, this trial completed in June 2016. Results were published after peer review (Chamberlain et al., 2020). The drug appeared safe, and its plasma pharmacokinetics predicted brain concentrations sufficient to activate PPAR delta. It improved blood markers of insulin sensitivity, and increased brain glucose metabolism in multiple brain regions. Some improvements in ADAS-Cog 11 were found, possibly related to ApoE genotype.

In May 2019, the company announced it had received NIA funding to begin a Phase 2 study called PIONEER (Prospective Therapy to Inhibit and Overcome Alzheimer’s Disease Neurodegeneration via Brain EnErgetics and Metabolism Restoration). Starting in March 2021, the trial enrolled 250 people with a clinical diagnosis of mild to moderate AD at 36 centers across the U.S. Participants were not required to have evidence of brain amyloid by PET or biomarkers. The study compared a six-month course of 15, 30, or 45 mg of T3D-959 to placebo. The primary outcomes were ADAS-Cog11, ADCS-Clinical Global Impression of Change, and safety. Secondary endpoints were the Digit Symbol Coding Test of executive function and plasma Aβ42/40. As of July 2022, the trial was 92 percent enrolled, according to an interim data presentation at the August 2022 AAIC. The drug appeared safe, with no serious adverse events reported, and none that caused treatment to be stopped. The trial was still blinded. Even so, the company claimed a possible treatment effect based on small improvements from baseline in both primary outcomes and on the secondary DCST, in the participants considered as one group including placebo recipients.

The trial finished in January 2023, and top-line results were presented at the October 2023 CTAD. The trial failed to meet its primary endpoints. Target engagement was inferred by changes in plasma glucose, insulin and lipid-related markers, and improvements in brain glucose metabolism. Biomarkers of plasma amyloid Aβ42/40 ratio and neurogranin both improved at the 30 mg but not the 45 mg dose. Only 42 percent of participants in this trial were ApoE4-positive. Only about 55 percent of participants were thought to be amyloid-positive, based on elevation of the plasma ptau217 to total tau ratio at baseline. A post hoc analysis of this subgroup indicated a two-point benefit in the ADAS-Cog for the 30 mg treatment, compared to placebo, but it lacked statistical power.

For trial details, see clinicaltrials.gov.

Last Updated: 22 Nov 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

PF-06648671

Tools

Back to the Top

Overview

Name: PF-06648671
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Pfizer

Background

PF-06648671 is a γ-secretase modulator Pfizer was developing for the treatment of Alzheimer's disease. No preclinical information is publicly available on this compound.

Findings

From December 2014 to March 2015, Pfizer conducted a single-ascending-dose study in 18 healthy volunteers to assess routine safety and pharmacokinetic parameters, as well as measures related to the pharmacodynamics of plasma Aβ40 and Aβ42.  

From May 2015 to October 2016, a second study enrolled 92 healthy elderly volunteers in Belgium for a two-week, once-daily regimen of ascending doses ranging from 4 to 100 mg of PF-06648671. It aimed to determine the maximum tolerated dose and assess drug interactions with the cytochrome P450 probe midalozam at that dose. Besides safety, pharmacokinetic, and pharmacodynamic measures of PF-06648671 in blood, CSF, and urine, additional outcome measures assessed the change from baseline in CSF concentrations of Aβ37, 38, 40, 42. 

From October 2015 to March 2016, Pfizer enrolled 22 healthy volunteers in California to characterize the effect of single doses on the dynamics of CSF Aβ37, 38, 40, 42, and Aβ total concentration using serial sampling of spinal fluid over 36 hours. This study used doses of 300 mg and lower. In the fall of 2016, Pfizer conducted a small drug interaction study with the CYP3A4 inhibitor itroconazole in 12 healthy volunteers.

At the 2016 AAIC conference, Pfizer reported that this γ-secretase modulator was safe and well-tolerated at single doses of up to 360 mg and dose-dependently lowered plasma Aβ40 and Aβ42 (Qiu et al., 2016). At the 2017 AAIC conference, Pfizer scientists reported that, as modeled, PF-06648671 shifted APP cleavage away from Aβ42 production and toward Aβ37 and Aβ38 production in CSF, without changing total Aβ levels (Ahn et al., 2017).

In January 2018, Pfizer announced it was discontinuing research and development in neurology, including this compound. Trial results were subsquently published (Ahn et al., 2020).

For all trial listings on this drug, see clinicaltrials.gov.

Last Updated: 06 Jan 2022

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

TDP-43 (WT) (Elliott)

Synonyms: WT TDP-43 (line 4)

Tools

Back to the Top

Species: Mouse
Genes: TARDBP
Modification: TARDBP: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis
Strain Name: B6SJL-Tg(Prnp-TARDBP)4Jlel/J

Summary

This model was among the first wave of TDP-43 transgenics (Stallings et al., 2010). It was originally created on a hybrid background (B6SJLF and CD1), and later developed into a  B6SJL line distributed by The Jackson Lab (see below). The characterization data on this page refer to the original hybrid mice unless otherwise noted.

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

  • Cortical Neuron Loss
  • Lower Motor Neuron Loss

No Data

  • NMJ Abnormalities
  • Gliosis

Cortical Neuron Loss

Not observed.

Lower Motor Neuron Loss

Not observed.

Cytoplasmic Inclusions

Cytoplasmic ubiquitin-positive inclusions in skeletal muscle cells. Some TDP-43 inclusions, too.

Gliosis

No data.

NMJ Abnormalities

No data.

Muscle Atrophy

An analysis of the quadriceps muscle, showed signs of myopathy, including variable muscle fiber size and disorganization of the muscle architecture.

Motor Impairment

Progressive motor impairment starting with external rotation of one hind limb followed by bilateral weakness and low muscle tone. Variable penetrance of this phenotype.

Body Weight

Progressive weight loss.

Premature Death

The mean survival of hemizygous mice was 109 days (it is not clear if this value represents males, females, or both).

Last Updated: 05 Jun 2019

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

TDP-43 (M337V) (Mt-TAR6/6)

Tools

Back to the Top

Species: Mouse
Genes: TARDBP
Modification: TARDBP: Transgenic
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
Strain Name: N/A

Summary

These mice, which express mutant human TDP-43 in postnatal neurons, quickly develop several ALS-related phenotypes, including motor impairment, upper motor neuron loss, and premature death (Janssens et al., 2013). The “Mt” in the name of this model stands for “mutant,” and refers to the fact that the transgene contains the M337V mutation associated with familial ALS.

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

No Data

  • NMJ Abnormalities
  • Muscle Atrophy

Cortical Neuron Loss

Severe neuronal loss in all CA regions of the hippocampus of homozygous mice. Neuronal loss was also observed in layer V cortical neurons and thalamic neurons.

Lower Motor Neuron Loss

Neuronal loss was observed in the spinal cords of homozygous mice.

Cytoplasmic Inclusions

Some homozygous mice developed cytoplasmic inclusions in layer V cortical neurons. These were often, but not always, ubiquitin–positive. They were not universally observed, even in end-stage mice.

Gliosis

Elevated astrogliosis and microgliosis compared with non-Tg controls, especially in the motor cortex and spinal cord. Gliosis in the hippocampus was seen at end stage.

NMJ Abnormalities

Unknown.

Muscle Atrophy

Unknown.

Motor Impairment

Motor impairment developed quickly, by 11 days of age in homozygous mice, starting with an abnormal clasping reflex. They also develop a hunched posture, muscle twitches, and reduced mobility. Paralysis developed within days, leading to death. Hemizygotes do not develop motor symptoms until about one year of age, and impairment varied from mouse to mouse.

Body Weight

Early postnatal growth retardation in homozygous mice. By day 17 their average body weight is about half that of non-Tg controls.

Premature Death

Homozygous mice survived an average of just 17 days. In contrast, hemizygous Mt-TAR6 mice lived up to 24 months (average survival ~16.4 months).

Last Updated: 06 Mar 2018

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Subscribe to ALZFORUM RSS