Research Models

hCR1 KI on APOE4/Trem2

Tools

Back to the Top

Species: Mouse
Genes: Cr2, CR1, CR2, APOE, Trem2
Modification: Cr2: Knock-Out; CR1: Knock-In; CR2: Knock-In; APOE: Knock-In; Trem2: Knock-In
Disease Relevance: Alzheimer's Disease
Strain Name: B6(SJL)-Cr2tm1(CR2,CR1)How Apoetm1.1(APOE*4)Adiuj Trem2em1Adiuj/J

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

No Data

  • Plaques
  • Tangles
  • Gliosis
  • Synaptic Loss
  • Changes in LTP/LTD
  • Cognitive Impairment

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Gliosis

No data.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 04 Dec 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

Kif21b*T82T/APOE4/Trem2*R47H

Tools

Back to the Top

Species: Mouse
Genes: Kif21b, APOE, Trem2
Modification: Kif21b: Knock-In; APOE: Knock-In; Trem2: Knock-In
Disease Relevance: Alzheimer's Disease
Strain Name: B6(SJL)-Kif21bem1Adiuj Apoetm1.1(APOE*4)Adiuj Trem2em1Adiuj/J

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

No Data

  • Plaques
  • Tangles
  • Neuronal Loss
  • Gliosis
  • Synaptic Loss
  • Changes in LTP/LTD
  • Cognitive Impairment

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Neuronal Loss

No data.

Gliosis

No data.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 04 Dec 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

Mthfr*C677T/APOE4/Trem2*R47H

Tools

Back to the Top

Species: Mouse
Genes: Mthfr, APOE, Trem2
Modification: Mthfr: Knock-In; APOE: Knock-In; Trem2: Knock-In
Disease Relevance: Alzheimer's Disease
Strain Name: B6(SJL)-Mthfrem1Adiuj Apoetm1.1(APOE*4)Adiuj Trem2em1Adiuj/J

Summary

The epsilon-4 allele of Apoliporotein E and the R47H variant of TREM2 have each been found to confer an approximately threefold increased risk for Alzheimer’s disease in humans heterozygous for either allele. The C677T variant of MTHFR (methylenetetrahydrofolate reductase), which increases plasma homocysteine levels, is also associated with an increased risk of AD (Peng et al., 2015).

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

  • Plaques
  • Tangles
  • Neuronal Loss
  • Gliosis
  • Synaptic Loss
  • Changes in LTP/LTD
  • Cognitive Impairment

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Neuronal Loss

No data.

Gliosis

No data.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 04 Dec 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

Remternetug

Tools

Back to the Top

Overview

Name: Remternetug
Synonyms: LY3372993, N3pG-Abeta mAb
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 3)
Company: Eli Lilly & Co.

Background

Remternetug, previously LY3372993, is an investigational monoclonal antibody. Lilly's development pipeline lists it as an N3pG-Aβ mAb, suggesting it recognizes a pyroglutamated form of Aβ that is aggregated in amyloid plaques. LY3372993 is a follow-on to donanemab, the company's FDA-approved antibody also against pyroglutamated Aβ.

Findings

In November 2018, Lilly started a Phase 1 trial with the intention to compare LY3372993 to placebo in 100 healthy adults and people with MCI due to Alzheimer's disease or mild to moderate AD. The study delivered intravenous infusions of single or multiple escalating doses of LY3372993, and assessed adverse events, LY3372993 exposure in blood, and its ability to affect brain amyloid as measured by florbetapir PET. This study was set to run at eight U.S. centers until October 2020, but in April 2019, Lilly decided to terminate the trial after testing 36 healthy adults, and never enrolled any participants with AD. Study results were not made public.

In July 2020, Lilly began a separate Phase 1 study in people with AD. Thirty participants with clinically diagnosed mild cognitive impairment due to AD, or AD dementia, are receiving multiple escalating doses of antibody, or placebo, over nine months. The primary outcome is the number of serious adverse events; secondary outcomes include pharmacokinetics and change in brain amyloid from baseline to week 25. In 2021, the company added a second part to this trial, enrolling an additional 32 healthy adults of first-generation Japanese origin to receive a single infusion of antibody or placebo. Through 2021 and 2022, enrollment was adjusted upward twice to a final of 224, the treatment period increased to 61 weeks, and the study added subcutaneous dosing. Conducted at 10 U.S. sites and three in Japan, the trial will run through November 2024.

The company presented interim Phase 1 data at the March-April 2023 AD/PD Conference in Gothenburg, Sweden (Apr 2023 conference news). They reported on 41 participants who received 250, 700, 1,400, or 2,800 mg of remternetug, monthly, or placebo, for six months. One cohort was titrated from 700 to 1,400 mg. After six months, plaque was dose-dependently reduced in all cohorts. On 2,800 mg, every participant dropped below the amyloid positivity threshold within three months. Safety data remains blinded, but overall there were 10 ARIA-E and seven ARIA-H cases, without obvious dose correlation. All the ARIA-E cases occurred in APOE4 carriers; one was symptomatic. Neither antidrug antibodies nor systemic infusion reactions were identified.

In August 2022, Lilly started a Phase 3 called TRAILRUNNER-ALZ1. It was initially going to randomize 400 people with early symptomatic AD to receive antibody or placebo for one year, with dosing by either intravenous infusion or subcutaneous injection. The primary outcome is percentage of patients whose amyloid plaques are cleared by the end of treatment period. Secondary outcomes further measure amyloid clearance, pharmacokinetics, and anti-drug antibodies. The trial includes a year-long blinded extension, where remternetug recipients will cross over to placebo, and placebo recipients to drug. An additional safety cohort of 400 patients will receive open-label intravenous remternetug for one year. In 2023, enrollment in the placebo-controlled study was increased to 600. The safety cohort expanded to 640, and added subcutaneous dosing. Another expansion brought the safety cohort to 974. The study is running at 73 sites in the U.S. and three in Japan. The blinded portion of the study finished in June 2024. Study completion is expected in March 2026.

On May 7, 2024, the Dominantly Inherited Alzheimer Network announced that remternetug would be tested in the Knight Family DIAN-TU Primary Prevention Trial (press release). According to a presentation at the 2024 AAIC, the trial will enroll 240 people as young as 18, and will include mutation carriers and noncarriers (Aug 2024 news). Participants will get remternetug four times annually for two years, via subcutaneous injection. The primary outcome is amyloid accumulation, with fluid biomarkers as secondaries. A four-year open-label extension will be offered to mutation carriers, and include cognition as a secondary outcome. Enrollment began in November 2024, with completion anticipated in 2034. This study was originally planned to begin in December 2022 and test Roche’s gantenerumab, but its launch was delayed when development of that antibody stopped.

In October 2024, a Phase 3 trial began to test remternetug in early Alzheimer’s patients. The TRAILRUNNER-ALZ 3 study plans to enroll 1,200 participants for 18 months of home treatment with subcutaneous injections, followed by a blinded observation period, against a primary endpoint of time to progression on the Clinical Dementia Rating (Nov 2024 conference news). Secondary outcomes include the CDR-SB, and a battery of cognitive, behavioral, and functional tests, serum antibody concentrations, and adverse events. Participants must have plasma pTau217 consistent with amyloid pathology, and no or minimal cognitive or functional impairment. The trial includes an amyloid and tau PET substudy, and an open-label extension. It is planned to run until 2029.

For all trials of LY3372993, see clinicaltrials.gov.

Last Updated: 08 Jan 2025

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-TAR6/6)

Synonyms: WT-TAR6/6, 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

This mouse model of ALS expresses wild-type human TDP-43 in postnatal neurons. Mice homozygous for the human TARDP transgene are referred to as TAR6/6, while mice hemizygous for the transgene are called TAR6. This description refers to homozygous, TAR6/6, mice. These mice exhibit neuropathological features of ALS, including elevated amounts of cytoplasmic and insoluble TDP-43, astrogliosis and microgliosis, and loss of motor neurons in the cortex and spinal cord; motor impairment; and early death.

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

Approximate 15 percent loss of layer V neurons in motor cortex at 6 months.

Lower Motor Neuron Loss

Spinal motor neuron loss observed at 3 months, with approximately 10 percent fewer anterior horn neurons in lumbosacral regions at 6 months, compared with non-transgenic mice.

Cytoplasmic Inclusions

Inclusions containing phosphorylated TDP-43 rarely observed in the nuclei and cytoplasm of spinal neurons.

Gliosis

Microgliosis in cortex and spinal cord prominent at 6 months; astrogliosis in cortex and spinal cord apparent at 1.5 months.

NMJ Abnormalities

No data.

Muscle Atrophy

Muscle wasting, particularly in flanks.

Motor Impairment

Progressive motor impairment; abnormal hind limb reflexes observed as early as 1.5 months.

Body Weight

TAR6/6 mice had lower body weights than non-transgenic mice between 3.25 and 3.75 months age, but the two genotypes were similar at younger and older ages (at least until 4.25 months).

Premature Death

Average survival is 6.7 months.

Last Updated: 21 Sep 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

AAV-GFP–(GR)100

Synonyms: GFP–(GR)100

Tools

Back to the Top

Species: Mouse
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
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

  • Lower Motor Neuron Loss
  • Body Weight

No Data

  • NMJ Abnormalities
  • Muscle Atrophy
  • Premature Death

Cortical Neuron Loss

Fewer cortical neurons than controls, observed as early as 6 weeks of age.

Lower Motor Neuron Loss

Not observed up to 6 months of age.

Cytoplasmic Inclusions

Very rare TDP-43 inclusions, observed at 6 months.

Gliosis

Astrogliosis and microgliosis in cortex and hippocampus, observed at 6 weeks.

NMJ Abnormalities

No data.

Muscle Atrophy

No data.

Motor Impairment

Progressive motor deficits, seen in open-field, rotarod, rod-walk, and wire-hang tests.

Body Weight

Normal at 6 months of age.

Premature Death

No data.

Last Updated: 21 Sep 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

AAV-GFP-(GA)50

Synonyms: GFP-(GA)50

Tools

Back to the Top

Species: Mouse
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
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

No Data

  • Lower Motor Neuron Loss
  • NMJ Abnormalities
  • Muscle Atrophy
  • Premature Death

Cortical Neuron Loss

At 6 months, neuron loss observed in cortex, including layer V of motor cortex.

Lower Motor Neuron Loss

No data.

Cytoplasmic Inclusions

Neuronal poly(GA)- and ubiquitin-positive inclusions already present at 4-6 weeks; rare TDP-43-positive inclusions.

Gliosis

Astrogliosis, but not microgliosis, in cortex and hippocampus, seen at 6 months.

NMJ Abnormalities

No data.

Muscle Atrophy

No data.

Motor Impairment

Deficits in tail-suspension and rotarod tests, seen at 6 months.

Body Weight

Decreased body weight in males, but not females, seen at 6 months.

Premature Death

No data.

Last Updated: 21 Sep 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

Trem2 R47H KI (Haass)

Synonyms: R47H ki

Tools

Back to the Top

Species: Mouse
Genes: Trem2
Modification: Trem2: Knock-In
Disease Relevance: Alzheimer's Disease
Strain Name: Trem2em2 Bwef

R47H is a rare variant in TREM2 that triples the risk of Alzheimer’s disease in heterozygous carriers. To create a mouse model carrying a single copy of R47H Trem2 under the control of its natural regulatory elements, CRISPR/Cas9 was used to introduce the R47H variant into the endogenous mouse Trem2 gene (Xiang et al., 2018).

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

  • Plaques
  • Tangles
  • Neuronal Loss
  • Gliosis
  • Synaptic Loss
  • Changes in LTP/LTD
  • Cognitive Impairment

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Neuronal Loss

No data.

Gliosis

No data.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 17 Sep 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

McGill-R-Thy1-APP

Tools

Back to the Top

Species: Rat
Genes: APP
Modification: APP: Transgenic
Disease Relevance: Alzheimer's Disease
Strain Name: N/A

Summary

The McGill-R-Thy1-APP transgenic rat expresses human APP751 with the Swedish and Indiana mutations, under the control of the murine Thy1.2 promoter. These rats are among the most extensively studied of the existing APP transgenic rat lines. Homozygous animals show age-dependent accumulation of amyloid plaques, gliosis, cholinergic synapse loss, and cognitive impairment.

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

  • Tangles

Plaques

Amyloid plaques present in homozygotes, appearing in hippocampus at 6 months and cortex at 13 months. Plaques are generally absent in hemizygotes.

Tangles

No data.

Synaptic Loss

Reduction in cholinergic synaptic boutons seen at 20 months in homozygous transgenic rats.

Neuronal Loss

A 22 percent reduction in the number of neurons was seen in the subiculum of homozygous transgenic rats at 18 months.

Gliosis

Microgliosis and astrogliosis observed in homozyogotes.

Changes in LTP/LTD

Impairments in long-term potentiation in CA1 by 3.5 months of age.

Cognitive Impairment

Deficits in Morris water maze and fear conditioning test are apparent by 3 months of age in both hemizygous and homozygous transgenic rats.

Last Updated: 31 Aug 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

TgF344-AD

Tools

Back to the Top

Species: Rat
Genes: APP, PSEN1
Mutations: PSEN1: deltaE9
Modification: APP: Transgenic; PSEN1: Transgenic
Disease Relevance: Alzheimer's Disease
Strain Name: N/A

Summary

TgF344-AD rats express human APP with the Swedish mutation and human PSEN1 with the Δ exon 9 mutation. Both transgenes are driven by the mouse prion promoter (Cohen et al., 2013).

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

  • Synaptic Loss
  • Changes in LTP/LTD

Plaques

Age-dependent accumulation of amyloid plaques in hippocampus and cortex between 6 and 26 months of age.

Tangles

Structures similar in appearance to neurofibrillary tangles revealed by Gallyas staining and immunostaining using an antibody directed against phospo-tau.

Synaptic Loss

No data.

Neuronal Loss

Approximate 40 percent loss of neurons in hippocampus and cortex by 16 months.

Gliosis

Microgliosis and astrogliosis are apparent by 6 months.

Changes in LTP/LTD

No data.

Cognitive Impairment

Deficits in reversal learning in the Morris water maze apparent by 6 months.

Last Updated: 10 Aug 2023

COMMENTS / QUESTIONS

  1. This work deserves a lot of attention. One thing that is unclear is the definition of tauopathy.

    View all comments by Takaomi Saido

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