Research Models

MAPT knock-in

Synonyms: MAPT KI, hTau KI

Tools

Back to the Top

Species: Mouse
Genes: MAPT
Modification: MAPT: Knock-In
Disease Relevance: Alzheimer's Disease, Other Tauopathy
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

  • Plaques
  • Tangles
  • Neuronal Loss
  • Gliosis
  • Cognitive Impairment

No Data

  • Synaptic Loss
  • Changes in LTP/LTD

Plaques

No amyloid plaques at 24 months of age.

Tangles

No neurofibrillary tangles at 24 months of age.

Synaptic Loss

No data.

Neuronal Loss

Neurodegeneration not apparent up to 2 years of age.

Gliosis

No astrogliosis or microgliosis observed at 24 months.

Changes in LTP/LTD

No data.

Cognitive Impairment

At 12 months of age, MAPT knock-in mice perform similarly to wild-type mice in the Y-maze test of working memory (only males tested).

Last Updated: 13 Aug 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

LRRK2 G2019S KI Mouse

Tools

Back to the Top

Species: Mouse
Genes: LRRK2
Modification: LRRK2: Knock-In
Disease Relevance: Parkinson's Disease
Strain Name: C57BL/6-Lrrk2tm4.1Arte

This constitutive knock-in (KI) mouse model was generated by introducing the LRRK2 G2019S point mutation into exon 41 of the mouse LRRK2 gene (Matikainen-Ankney et al., 2016). Homozygous mutant mice appear grossly normal. They generate litters comparable in size to wild-type animals, and grow at a normal rate.

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

  • Dopamine Deficiency
  • Neuronal Loss

No Data

  • α-synuclein Inclusions
  • Neuroinflammation
  • Mitochondrial Abnormalities

Neuronal Loss

The cytoarchitecture of the neocortex, striatum, hippocampus, and elsewhere is normal in Nissl-stained brain sections of 3-4 month-old mice, and striatal levels of tyrosine hydroxylase are similar to those of controls at P21. In another study, tyrosine hydroxylase levels are reduced in the striatum and midbrain at 2 months of age.

Dopamine Deficiency

Striatal dopamine levels do not differ at 2 months of age, and neither do tyrosine hydroxylase levels in the substantia nigra.

α-synuclein Inclusions

No data.

Neuroinflammation

No data.

Mitochondrial Abnormalities

No data.

Motor Impairment

A battery of motor tests revealed no baseline deficits at 3-4, 12-13, and 18-19 months of age. However, an increased locomotor response after amphetamine challenge is observed at 18 months. Motor defects are exacerbated following a manganese stressor.

Non-Motor Impairment

Altered responses to social-defeat stress (males, 3-4 mos) which correlated with changes in striatal plasticity and intrinsic membrane excitability. Attention deficits, slower information processing, impaired goal-directed learning in 2-6-month-old male KI mice, but cognitive flexibility and novel objective recognition are intact (2-6 mos). Perturbed sleep behavior at 8-10 mos.

Last Updated: 20 Dec 2023

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

CamKII;(GR)80

Synonyms: CamKII-tTA;(GR)80 (line 16), CamKII;(GR)80

Tools

Back to the Top

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

A G4C2 hexanucleotide repeat expansion in chromosome 9 open reading frame 72 (C9ORF72) is the most frequent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This repeat expansion encodes five dipeptide repeat proteins that accumulate in ALS/FTD: glycine-arginine (GR), proline-arginine (PR), glycine-alanine (GA), proline-alanine (PA), and glycine-proline (GA). CamKII;(GR)80 mice were created to study the effects of poly(GR).

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

  • Motor Impairment
  • Cytoplasmic Inclusions
  • Body Weight

No Data

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

Cortical Neuron Loss

Neuron loss in the cortex, beginning between 3 and 6 months of age.

Lower Motor Neuron Loss

No data, but note that the transgene is not expressed in these neurons.

Cytoplasmic Inclusions

No TDP-43 inclusions seen in mice studied up to 8 months of age.

Gliosis

Microgliosis and astrogliosis evident at 6 and 9 months, respectively.

NMJ Abnormalities

No data.

Muscle Atrophy

No data.

Motor Impairment

No difference between CamKII;(GR)80 mice and CamKII-tTA control mice up to 9 months of age.

Body Weight

No difference between CamKII;(GR)80 mice and CamKII-tTA control mice up to 11 months of age.

Premature Death

No data.

Last Updated: 01 Jul 2019

COMMENTS / QUESTIONS

  1. This paper nicely shows that expression of a poly(GR) in all the layers of the cortex under the control of a CamKII promotor can induce an ALS/FTD-related phenotype. A major advantage of this study is that the poly(GR) is expressed at a relatively low level and that phenotypes induced by huge overexpression of the transgene are avoided.

    Another very interesting characteristic of the system used in this study is that the expression of poly(GR) can be reduced by feeding the mice with doxycycline. Interestingly, behavioral and even the cellular phenotypes can be significantly reversed when the expression of poly-GR is lowered. This is fascinating and has far-reaching consequences. It strongly indicates that therapeutic strategies lowering the expression of toxic dipeptide-repeat proteins can reverse disease phenotypes, even after disease onset. This reversal of the phenotype can be considered as a very interesting new insight, in addition to the systematic characterization of the underlying disease mechanism, which is related to mitochondrial dysfunction.

    View all comments by Ludo Van Den Bosch
  2. This paper describes an interesting new poly(GR)-expressing mouse model. We will need a range of different models to ultimately understand the role of each DPR and their contribution to C9ORF72-repeat pathology, so this is a welcome addition.

    The most striking finding is that neuronal loss and synaptic dysfunction are identified even though GR is not detectable by immunostaining until six to eight months of age (although it can be detected earlier with ELISA). This suggests relatively low levels of likely soluble GR are sufficient to induce neurodegeneration.

    The paper also links poly(GR) to an early defect in mitochondrial function, which could explain the later development of DNA damage. Many pathways have been implicated in C9ORF72-repeat-induced neurodegeneration, and mitochondrial dysfunction should now be added as a new avenue for further investigation. Consistent with the previously reported AAV poly(GR) mouse model, no TDP-43 pathology was observed, and so the link between DPRs and TDP-43 is still an outstanding question.

    View all comments by Adrian Isaacs
  3. This is a very nice addition to the building literature on mechanisms of toxicity from DPR expression in animal models. It brings to light a surprisingly specific mechanism, whereby poly(GR) alters mitochondrial ATP5A1, further linking mitochondrial dynamics and dysfunction to neurodegeneration and providing a potential therapeutic avenue.

    Challenges going forward for the field include interpreting this mechanism of toxicity in the context of the numerous other toxicities of the different DPRs, and determining whether some are more important than others, and where and when they are happening in human disease. 

    Regardless, it provides further support for the idea that diminishing gain-of-function products of the C9ORF72 repeat could have a therapeutic effect, and many of these strategies are in preclinical or clinical phase testing.

    View all comments by Robert Baloh
  4. This work provides an additional mouse model and research tool for the C9 ALS/FTD community. Because C9 ALS/FTD disease is a complex disease, with the expression of two mutant transcripts (sense and antisense) and six dipeptide repeat-containing proteins, which are produced by repeat-associated non-AUG (RAN) translation, this polyGR-expressing mouse model is a simplified model for studying the potential contribution of polyGR to disease pathology.

    The paper provides a compelling data set showing that moderate expression of polyGR causes mitochondrial dysfunction that is linked to reductions in Atp5a1. In the future, it will be interesting to test if strategies described to reverse GR toxicity improve phenotypes in mouse models that express the repeat expansion and produce the multiple types of RAN proteins found in patients. It will also be of interest to test if Atp5a1 levels are reduced in these G4C2 mouse models and also in C9 patient tissues.

    View all comments by Laura Ranum

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

C9ORF72(AAV)(G4C2)149

Synonyms: (G4C2)149, 149-repeat mice

Tools

Back to the Top

Species: Mouse
Genes: C9orf72
Mutations: Hexanucleotide repeat in C9ORF72
Modification: C9orf72: Virus
Disease Relevance: Amyotrophic Lateral Sclerosis, Frontotemporal Dementia
Strain Name: N/A

A (G4C2) hexanucleotide repeat expansion in chromosome 9 open reading frame 72 (C9ORF72) is the most frequent genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis. Bidirectional transcription of this expansion yields sense (G4C2) and antisense (G2C4) repeat RNAs, which can then be translated—via repeat-associated non-ATG (RAN) translation—into a series of dipeptide repeat proteins.

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
  • Body Weight
  • Premature Death

Cortical Neuron Loss

Cortical neuron loss by 6 months.

Lower Motor Neuron Loss

No data.

Cytoplasmic Inclusions

Cytoplasmic inclusions containing dipeptide repeat proteins (DPRs) derived from sense RNA seen in the cortex, hippocampus, cerebellum, and spinal cord; inclusions containing DPRs produced from antisense transcripts seen in the cortex and occasionally in the hippocampus.

Gliosis

Astrogliosis in the cortex by 3 months.

NMJ Abnormalities

No data.

Muscle Atrophy

No data.

Motor Impairment

Deficits in the hanging wire test emerge between 3 and 6 months.

Body Weight

No data.

Premature Death

No data.

Last Updated: 28 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

Sensory Stimulation Systems

Tools

Back to the Top

Overview

Name: Sensory Stimulation Systems
Synonyms: GENUS, Gamma entrainment using sensory stimuli, GammaSense Stimulation
Therapy Type: Combination, Procedural Intervention
Target Type: Amyloid-Related (timeline), Inflammation (timeline), Other (timeline)
Condition(s): Alzheimer's Disease, Parkinson's Disease, Down's Syndrome
U.S. FDA Status: Alzheimer's Disease (Phase 3), Parkinson's Disease (Not Regulated), Down's Syndrome (Not Regulated)
Company: Cognito Therapeutics, Inc.

Background

Sensory Stimulation Systems, and GENUS, refer to a noninvasive procedure, i.e., a "digital therapeutic," being developed by Cognito Therapeutics. The company developed a wearable GammaSense Stimulation device. It pairs headphones and opaque glasses to deliver light and sound stimuli at gamma band frequency in order to entrain gamma oscillations in the brain. An LED light flashing at 40 Hz for one hour was reported to boost gamma power in the visual (V1), prefrontal, and somatosensory cortices, and hippocampal CA1; it also increases synchronization between V1 and the other regions. A combination of light and sound at 40 Hz has been shown to strengthen gamma oscillations throughout the auditory cortex, hippocampus, and medial prefrontal cortex. Gamma oscillations play a role in sensory processing, cognition, and memory consolidation, and are weakened in AD.

In 5XFAD, APPPS1, and wild-type mice, one hour of light-based GENUS reportedly halved Aβ levels, and one week of GENUS in 6-month-old 5XFAD mice cut amyloid plaque load by two-thirds. In TauP301S, this regimen cut phosphorylated tau by 40 percent (Iaccarino et al., 2016).

Neuron numbers were maintained in TauP301S mice for three weeks with GENUS starting at 7.5 months of age, when neurodegeneration begins in this model. In the inducible p25 neurodegeneration mouse model, six weeks of GENUS preserved neuron number and cortical thickness. In both models, GENUS maintained neuronal expression of genes affecting synaptic transmission and vesicle trafficking, protected DNA, increased the density of mushroom spines, and improved performance in the Morris water maze. In all models tested, GENUS lowered microgliosis and inflammatory gene expression, while increasing internalization of Aβ (Adaikkan et al., 2019). The combination of light and sound at 40 Hz in 6-month-old 5XFAD mice had similar effects to light alone, but reached more brain areas (Martorell et al., 2019).

In other studies, 40 Hz light flicker or ultrasound was reported to improve cognitive function, enhance presynaptic plasticity, and activate microglia to reduce Aβ plaque load (Singer et al., 2018Park et al., 2020; Zheng et al., 2020; Bobola et al., 2020). A separate study linked acoustic stimulation to lowered amyloid plaques and increased microglial numbers in the hippocampi of 5XFAD mice (Lee et al., 2018). Multisensory 40 Hz stimulation was reported to increase glymphatic clearance of Aβ in the same model (Murdock et al., 2024), and to restore hippocampal synaptic potentiation in rat models (Yang et al., 2023). Other reported effects of 40 Hz light flicker in AD preclinical models include increased expression of cytokines that regulate microglia (Garza et al., 2020; Prichard et al., 2023), normalization of circadian rhythms, altered APP processing, and improved effects of exercise on cognition (Yao et al., 2020; Shen et al., 2022; Park et al., 2022).

In one study using 40 Hz optogenetic stimulation, hippocampal gamma waves and spatial memory changed, but plaque load did not. In another, plaques grew; memory was not assessed (Etter et al., 2019Wilson et al., 2020). In other negative studies, native gamma oscillation entrainment, amyloid lowering, and microglial activation were not replicated (Soula et al., 2023; Yang and Lai, 2023). Transgenic AD rats increased their activity, but did not improve cognition, after light and sound stimulation (Bentley and Broussad, 2024).

For reviews, see McDermott et al., 2018Adaikkan and Tsai, 2020Manippa et al., 2022; Blanco-Duque et al., 2024).

Devices that offer 40 Hz light and sound stimulation are sold directly to consumers. Evidence for their claimed uses is sparse.

Findings

In a brief pilot study conducted in Europe, without Cognito Therapeutics, 10 days of 40 Hz light therapy, administered for two hours per day, did not affect amyloid load, as measured by PiB PET, in 10 AD patients (Ismail et al., 2018).

In April 2018, Cognito Therapeutics began a Phase 1 trial called Overture to evaluate its CogTx-001 GammaSense stimulation device in 60 people with AD or mild cognitive impairment due to AD. Devices were calibrated for each participant and EEG-verified to elicit 40 Hz steady-state gamma oscillations. Participants received one hour of combined 40 Hz auditory and visual stimulation, or a sham treatment, daily for six months at home, with a follow-up visit one month later to assess safety. The primary outcome was ADAS-Cog, CDR-SB, and MADCOMS scores, with ADCS-ADL and MMSE and amyloid imaging as secondary outcomes. The trial took place at five sites in the U.S. and ran until August 2021.

In March 2021, results were presented at AD/PD. Forty-six participants received active stimulation, 28 sham. There were no serious adverse events, but more participants on active stimulation complained of tinnitus. In each group, 28 percent of participants dropped out. The trial missed all three primary outcomes. Of the 53 people who finished the six-month regimen, 44 continued to a one-year, open-label extension (Apr 2021 conference news). Published results on the first 22 participants reported a decrease in nighttime activity. Active treatment was reported to result in stabilization on the ADCS-ADL, compared to a decline in the sham group (Cimenser et al., 2021). Complete trial results were published, and showed no statistically significant changes in any of the primary or secondary endpoints, nor any change in amyloid levels on PET (Hajós et al., 2024). Additional analysis of MRI data suggested reduced white matter atrophy and myelin content loss, especially in the entorhinal cortex (Da et al., 2024). Results of the open-label extension were presented at the March 2024 AD/PD conference. Adherence was 80 percent, with no new adverse events of tinnitus or headache reported. ADCS-ADL scores were stable in the 15 continuously treated patients. The seven patients who started on placebo were reported to have shown a trend toward slowing of decline in the ADCS-ADL and loss of brain volume after switching to active treatment. Progression modeling implied a disease-modifying effect.

In May 2018, a second Phase 1 study called Etude began comparing dosing paradigms for auditory and visual stimulation with the GammaSense device in 20 people with AD or mild cognitive impairment due to AD. These included one hour once per day, one hour twice per day, one hour every other day, and 30 or 120 minutes twice per day. The primary outcomes were amyloid PET and safety; secondary, ADAS-Cog. The trial was to be completed by March 2022; no results have been reported.

From November 2018 to February 2020, a third study called Flicker evaluated the tolerability of a combined audio-visual stimulation for one hour per day in 10 people with mild cognitive impairment due to AD. Five participants received eight weeks of GammaSense stimulation; five received four weeks of no intervention followed by four weeks of intervention. There was no placebo/sham. This investigator-initiated trial was conducted at Emory University, and sponsored by Cognito. As reported at AD/PD 2021, the study first tested escalating brightness and loudness. Most people tolerated full-intensity stimulation. Overall, participants completed 95 percent of sessions. No severe treatment-related adverse events were reported. Some participants reported dizziness, headaches, or tinnitus; one person’s hearing loss worsened. In exploratory measures, EEG detected gamma entrainment in many brain areas. Gamma power was the same after four weeks treatment, and weakened after eight. Strengthened brain connectivity was detected at eight weeks between the posterior cingulate and precuneus, two regions in the default mode network that is disrupted in AD. Brain amyloid and CSF Aβ or tau did not change after eight weeks, although CSF levels of many cytokines changed (Apr 2021 conference news, He et al., 2021). Nine of the 10 participants chose to enter a one-year, open-label extension.

Two other studies of gamma entrainment were conducted at MIT, with results also presented at AD/PD (April 2021 conference news). These studies used an easel-mounted array of lights and speakers, not the GammaSense device. One began in April 2019 and involved 46 healthy adults and 26 older people with mild AD. EEG monitoring was used to optimize stimulation parameters during 30-60 minute sessions. The 40 Hz light/sound stimulation increased gamma waves in most brain areas, while triggering no headaches, vision or hearing changes, or seizures.

A second study began in August 2019 to evaluate the effects of daily stimulation in 15 people with mild AD. Participants were randomized to active or sham stimulation for one hour daily using the same light/speaker array at home for six or nine months. The primary outcomes measured device usage and adverse events. Other outcomes included cognition, function, neuropsychiatric symptoms, blood levels of amyloid, tau, and neurofilament light, EEG changes, structural and functional MRI, sleep time and efficiency, and physical activity. After the first period, all participants had the option to continue on active settings for six or nine months. In this study, data collection was paused at three months due to the COVID pandemic. Preliminary results found daily stimulation resulted in better sleep by one month. At three months, active treatment strengthened functional MRI connectivity in brain circuits involved in vision and memory, improved performance on a face-name-matching test, and may have slowed loss of hippocampal volume. Participants reportedly could not tell if they received active or sham stimulation (Apr 2021 conference news). Results of both studies were subsequently published (Chan et al., 2022).

Seven participants in these studies entered the open-label extension. At the 2024 AD/PD conference, results were reported on three late-onset and two early onset AD patients, who completed treatment for a total of 30 months. Induction of 40 Hz neural activity was sustained, but brain connectivity was reduced. The properties of circadian rhythms improved compared to baseline; hippocampal volume loss slowed compared to historical controls. In the late-onset patients only, cognitive scores stabilized, with a preservation of performance on the MMSE, CDR-SB compared to historical controls.

In January 2021, Cognito's GammaSense stimulation device received breakthrough status from the FDA. 

In July 2022, the University of Tennessee Medical Center in Knoxville started enrolling 20 people with Alzheimer's disease into an open-label study of an eight-week course of one hour per day of treatment with Cognito’s GammaSense device. The primary outcome assesses cortical visual processing; the study will run until December 2025.

In December 2022, Cognito Therapeutics began enrolling for a Phase 3 trial in 345 people with AD. They are to be randomized to either active or sham treatment lasting one hour per day for up to a year, using a sensory stimulation device called GS120. Primary outcomes are change from baseline at 12 months on the ADCS-ADL and on the combined statistical test (CST) of the ADCS-ADL and MMSE. In mid-2023, the trial increased enrollment to 530, and later added a one-year open-label extension. It is being run at 68 sites across the U.S., and is expected to be complete in 2025.

In December 2022, a new feasibility study began at MIT, enrolling 60 people with mild AD for six months of one hour daily, at-home stimulation, with a sham control. This study uses the easel-mounted GENUS device developed at MIT, and also will test a vibrating device that delivers 40 Hz tactile stimulation (see Suk et al., 2023). Primary outcomes are 40 Hz brain entrainment, safety, functional connectivity by fMRI, and AD-related inflammation biomarkers based on RNA sequencing in blood. Other outcomes include changes in the microbiome, sleep/wake patterns, and a cognitive testing battery. Participants will have the option of continuing the regimen for one year in an open-label extension. Completion is anticipated in September 2026.

In January 2024, MIT and MGH researchers began a prevention trial. It plans to enroll 50 cognitively normal people with cerebral amyloid deposits to a one-year, one-hour daily at-home treatment with the easel-mounted GENUS device, against a primary outcome of change in brain amyloid on PET. Other outcomes are to be change in tau-PET, structural and functional MRI, EEG, night-time activity, blood biomarkers of AD, and compliance. A substudy will assess CSF biomarkers of amyloid and tau. The study also plans to measure changes in CSF flow, white matter and myelination by diffusion MRI, neuropsychiatric symptoms, gamma oscillations, sleep, activity levels and adverse events. Completion is planned for May 2026. This trial offers a one-year, open-label extension.

At Emory University, a Parkinson’s trial is set to begin in August 2024. This study will enroll 28 PD patients who have problems walking, for six months treatment with a commercially available stimulation device from the Canadian company MindAlive Inc. Primary outcomes are compliance and adverse events. The study will also assess general symptoms and specifically freezing of gait, as well as CSF biomarkers of amyloid and inflammation after active or sham stimulation. 

MIT investigators are also running safety, feasibility, and optimization trials of GENUS in people with PD, and with Down's syndrome.

For details on trials of this approach, see clinicaltrials.gov.

Last Updated: 25 Jul 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Apabetalone

Tools

Back to the Top

Overview

Name: Apabetalone
Synonyms: RVX208, RVX000222
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): MCI-VaD
U.S. FDA Status: MCI-VaD (Phase 3)
Company: Resverlogix Corporation

Background

Apabetalone binds and inhibits bromodomain and extra-terminal domain (BET) proteins, preventing them from binding acetylated lysines in histones to activate gene transcription. In published papers, apabetalone is reported to increase expression of ApoA-1 and suppress genes associated with kidney disease and vascular calcification (McLure et al., 2013; Wasiak et al., 2018; Gilham et al., 2019).

Apabetalone also reportedly suppresses expression of complement proteins (Wasiak et al., 2017). In wild-type mice challenged with lipopolysaccharide, apabetalone suppressed pro-inflammatory cytokines, complement proteins, and endothelial cell adhesion markers (May 2019 conference news).

Findings

The Phase 3 BETonMACE trial evaluated apabetalone in 2,425 people with Type 2 diabetes and coronary artery disease on a primary outcome of number of major cardiovascular events. The trial included a cognitive substudy of 467 participants age 70 or older, who took the Montreal Cognitive Assessment (MoCA) at baseline, annually thereafter, and when the study ended (Ray et al., 2019). Its outcomes included whether apabetalone slowed cognitive decline over a two-year period. 

The trial was completed in November 2019. According to results presented at the 2019 CTAD conference in December, it missed its primary endpoint. Apabetalone reduced cardiovascular events compared to placebo, but the difference did not reach statistical significance. The main study results were subsequently published (Ray et al., 2020).

In the cognitive substudy, 469 participants had test results at baseline, one year and a final visit, which averaged 27 months.  They were split into three groups based on baseline MoCA scores, corresponding to cognitively normal (MoCA of 26 or higher), MCI (22-25), or MCI to AD (below 22). Treatment had no effect in the cognitively normal or MCI groups, whose MoCA scores declined slightly or remained stable.  The 97 participants in the MCI to AD subgroup improved an average of 3 points on the MoCA, compared to one point for placebo. The investigators plan to measure AD biomarkers in plasma samples from the study, to learn if a benefit occurred in AD or vascular dementia, or both (Dec 2019 conference news).

Apabetalone is also being evaluated in pulmonary artery hypertension, kidney failure, and Fabry disease. For all trials of apabetalone, see clinicaltrials.gov.

Last Updated: 30 Apr 2020

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Latozinemab

Tools

Back to the Top

Overview

Name: Latozinemab
Synonyms: AL001, GSK4527223
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Other (timeline)
Condition(s): Frontotemporal Dementia, Amyotrophic Lateral Sclerosis
U.S. FDA Status: Frontotemporal Dementia (Phase 3), Amyotrophic Lateral Sclerosis (Inactive)
Company: Alector, GlaxoSmithKline (GSK)

Background

AL001 is a recombinant human anti-human sortilin (SORT1) monoclonal IgG1 developed by Alector in partnership with Abbvie. In June 2018, this antibody received orphan drug designation from the U.S. FDA for the treatment of frontotemporal dementia.

Sortilin is a type I membrane glycoprotein in the vacuolar protein sorting 10 protein (Vps10p) family. Sortilin is abundantly expressed in the central nervous system. As a sorting receptor in the trans-Golgi network, sortilin is involved in neurotrophin signaling, lysosomal degradation, and APP metabolism. Sortilin regulates the endocytosis and degradation of progranulin, and this process has been implicated in FTD pathophysiology (Hu et al., 2010). Progranulin mutations are a leading cause of frontotemporal dementia, and are associated with 50 to 70 percent reduction in progranulin protein levels. Blocking sortilin with AL001 is a strategy to increase progranulin levels. 

The SORL1 gene is one of the strongest genetic risk factors for Alzheimer's disease, and is associated with frontotemporal dementia, as well (Rogaeva et al., 2007Pottier et al., 2012Holstege et al., 2017Raghavan et al., 2018Pallesen and Vægter et al., 2012Philtjens et al., 2018).

Targeting the sortilin-progranulin axis was reported to improve outcomes associated with progranulin insufficiency in preclinical models (Lee et al., 2013).

In cell assays, latozinemab blocked the interaction of sortilin with progranulin, reduced sortilin protein levels, and increased progranulin. In a mouse model of FTD-PGN, a rodent cross-reactive form of AL001 decreased sortilin levels in white blood cells, restored plasma proganulin to normal levels, and rescued a social behavior deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in blood cells, and increased plasma and CSF progranulin by two to three times (Kurnellas et al., 2023).

Findings

In September 2018, a first-in-human trial started enrolling 64 healthy adults and people with progranulin mutations causative for FTD at eight sites across North America and the U.K. Called INFRONT, the plan was to compare up to six single-ascending doses of AL001 intravenous infusion to placebo on outcomes including safety, pharmacokinetics, and exposure. The trial was completed in December 2019. At AAIC in July 2019, Alector reported results from 50 healthy volunteers and four FTD-GRN patients. According to the abstract and press release, AL001 infusion was “generally safe and well tolerated,” and resulted in a dose-dependent increase in progranulin levels in plasma and CSF in healthy adults and FTD patients. The press release notes a doubling of progranulin concentration in CSF of asymptomatic and symptomatic patients, restoring progranulin levels to the normal range. The results were published after peer review (Kurnellas et al., 2023).

Details of the complete cohort of 14 FTD-GRN mutation carriers were presented at AAIC July 2020. In six asymptomatic FTD-GRN mutation carriers, CSF progranulin levels were restored to normal 12 days after a single dose of 60 mg/kg. Among eight symptomatic patients, three doses of 30 mg/kg over four weeks raised progranulin into the normal range for at least eight weeks after the last dose. CSF markers of lysosomal function, inflammation, and gliosis were partially normalized. A non-significant 14 percent reduction in plasma neurofilament light chain (NfL) was observed eight weeks after dosing. Data on safety and progranulin elevation in symptomatic patients were subsequently published (Ward et al., 2024). Adverse events were mild to moderate. Some participants had elevated cholesterol or triglyceride levels. No patients dropped out due to side effects, and there were no deaths in the study.

In September 2019, recruitment began for INFRONT-2, an open-label, multi-dose Phase 2 study in people with progranulin or C9ORF72 mutations causative of FTD. This trial is assessing safety, tolerability, PK, and pharmacodynamics in 40 participants receiving 60 mg/kg monthly infusions for nearly two years. The study is running at 12 sites in North America, Europe, and the U.K. It was slated to end in 2021, but the date has been pushed back to early 2026. According to an AAIC 2020 presentation, by May 2020 the trial had enrolled 15 patients, including some rollovers from the Phase 1 study. The longest-treated patient had received six months of drug, which remained safe and well-tolerated. CSF progranulin reached normal levels within 10 days after the first dose in all participants, and stayed elevated with continued treatment. Due to the COVID-19 pandemic, two patients missed a dose, and some missed biomarker evaluations. A preliminary analysis based on this incomplete data indicated that plasma NfL declined in six of eight participants who received all scheduled infusions. In one patient, plasma NfL decreased during the Phase 1 trial, rebounded off drug, and decreased again when treatment recommenced in Phase 2.

At the July 2021 AAIC, Alector presented 12-month data from 12 symptomatic FTD-GRN patients in INFRONT-2. Participants maintained normal plasma and CSF progranulin levels, with no new safety issues. The company also reported normalization of lysosomal and inflammatory biomarkers, and stabilization of NfL in this group. A comparison with historical controls showed potential for slowing decline on exploratory clinical endpoints of brain atrophy and dementia (Aug 2021 conference news). A cohort of 10 symptomatic FTD-CORF72 patients posted similar results, according to a company presentation at the March 2022 AD/PD conference (press release).

In July 2020, Alector started a Phase 3, placebo-controlled trial. Called INFRONT-3, it with the aim of enrolling 180 people between 25 and 85 who are at risk of or have frontotemporal dementia due to heterozygous mutations in the progranulin gene. Participants are receiving a monthly intravenous infusion of AL001 60 mg/kg or placebo in a blinded fashion, or open-label AL001, for up to two years. The primary outcome is change in a combination of the Clinical Dementia Rating plus the NACC FTLD-Sum of Boxes, a measure adapted for frontotemporal dementia patients. Secondary clinical outcomes include standard measures of disease severity or improvement. The study will evaluate volumetric MRI, as well as CSF and blood pharmacodynamic biomarkers. It is taking place at 65 sites in North America, Australia, and Europe, and will run through 2025. In October 2023, the trial was declared fully enrolled at 101 symptomatic patients; an open-label continuation study of once-monthly 60 mg/kg latonizemab is recruiting (press release).

In July 2021, the company announced an agreement with GlaxoSmithKline to co-develop AL001 for FTD and ALS (press release). In September 2021, the first patient was enrolled in Phase 2 for ALS due to C9ORF72 mutations. The study planned to randomize 45 participants to six monthly infusions of AL001 or placebo, added to their current treatment. The primary endpoints were to be safety, tolerability, pharmacokinetics and pharmacodynamics, including plasma and CSF progranulin levels, with blood and CSF neurofilament light chain concentration as a secondary outcome. The study was terminated in October 2022, after enrolling 5 patients. The reason given was that Alector is considering a different study in ALS. The ALS program no longer appears in the company pipeline.

In February 2024, the FDA granted latozinemab breakthrough therapy designation, based on Phase 2 results (press release).

Alector has a related sortilin antibody, AL101, in Phase 2 for Alzheimer’s disease.

For all clinical trials of AL001, see clinicaltrials.gov.

Last Updated: 10 Jul 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

AL003

Tools

Back to the Top

Overview

Name: AL003
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Inflammation (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: AbbVie, Alector

Background

AL003 is a monoclonal antibody that was being co-developed by Alector and Abbvie. According to Alector, it counteracts the function of sialic acid binding immunoglobulin-like lectin 3, aka Siglec-3, an inhibitory microglial transmembrane receptor that reportedly interacts with TREM2 (Oct 2015 news). The gene encoding Siglec-3, CD33, has been associated with AD risk via a protective variant, expression levels, and alternative splicing (Estus et al., 2019Katsumata et al., 2019Bertram et al., 2008). CD33 is also expressed on peripheral blood monocytes.

Findings

In March 2019, AL003 entered the clinic with a 54-person trial being run at six sites in the United States and Melbourne, Australia (May 2019 conference news). The trial finished in May 2021, and results were presented at the November 2021 CTAD conference (poster). In a single-ascending-dose phase, 38 healthy participants received infusions of one of eight escalating doses. Outcomes included safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity. In healthy volunteers, the drug was well tolerated up to the 15 mg/kg dose. One person in the 30 mg/kg dose group developed hip inflammation, and dosing in the 60 mg/kg group was stopped after one person had a severe hypersensitivity reaction. Both required hospitalization and treatment with steroids. In the subsequent multiple-ascending-dose phase, 10 participants with AD received two doses of 15 mg/kg, four weeks apart; two received placebo. All finished the study with no serious drug-related adverse events. The half-life of the antibody after multiple dosing was approximately 15 hours, and CNS levels reached 0.1-0.2 percent of plasma. Treatment lowered CD33 expression on blood monocytes, and boosted soluble CD33 in CSF, indicating target engagement.

On June 30 2022, AbbVie terminated the AL003 collaboration, according to an Alector SEC filing. No reason was given, and Alector removed AL003 from its pipeline. The companies are continuing to co-develop the TREM2 antibody AL002.

For details, see clinicaltrials.gov.

Last Updated: 14 Jul 2022

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

AL002

Tools

Back to the Top

Overview

Name: AL002
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Inflammation (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: AbbVie, Alector

Background

This humanized monoclonal IgG1 antibody was being developed in a partnership between Alector and AbbVie. It binds the microglial receptor TREM2 and activates signaling, increasing phosphorylation of TREM2's downstream effector Syk, and inducing microglia proliferation. AL002a, a variant of AL002 that recognizes mouse TREM2, reportedly doubled the number of CD11b-positive microglia in cortex and hippocampus 72 hours after injection in APP/PS1 mice. Microglia expressed more pro-inflammatory and repair genes, and nearby amyloid deposits were nearly halved (Dec 2016 conference news). The antibody produced similar results in 5XFAD mice after 72 hours.

Repeated injections of 50 mg/kg AL002a into 4-month-old 5XFAD mice for 14 weeks reportedly raised the number of CD11b-positive microglia by 50 percent. The number of microglia around amyloid plaques doubled, and amyloid load was cut in half. Treated mice performed like wild type in the radial arm water maze and novel object recognition tasks (May 2019 conference news).

An anti-human TREM2 antibody, AL002c, was tested in 5XFAD mice carrying human TREM2 genes. Three months of weekly injection with 30 mg/kg AL002c activated microglia, reduced neurotoxicity and inflammatory signaling, and normalized behavior in an elevated maze. Treatment did not alter Aβ plaque load (news on Wang et al., 2020). AL002c is the preclinical variant of AL002.

Findings

In November 2018, AL002 started INVOKE, a Phase 1 trial at six sites in the U.S., Australia, and the U.K. In its single-ascending-dose phase, 69 healthy adults received a single infusion of one of nine doses between 0.003 to 60 mg/kg or placebo; in the multiple-ascending dose phase, 30 AD patients were enrolled in three dose cohorts. Outcomes included safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity. Some results of the single-dose portion of the study have been published (June 2020 news). The antibody was well tolerated, and no serious adverse events were noted up to 12 weeks after dosing. Treatment caused dose-dependent changes in two pharmacodynamic markers in CSF: soluble TREM2 was decreased two days after treatment, while a fragment of CSF1R, a receptor expressed only by microglia in the brain, was increased. The trial was completed in December 2020, and full trial results were subsequently published (Long et al., 2024). The half-life of AL002 in blood was 8 to 9 days, and CSF entry ranged from 0.17 to 0.28 percent after 13 days. The reduction in CSF sTREM2 persisted for 30 days in the highest dose group, and was accompanied by changes in other markers of microglia activity. The multiple dose study in AD patients was terminated early due to the COVID-19 pandemic. Pharmacokinetic and target engagement studies after multiple dosing were completed in non-human primates, and informed dose selection for Phase 2.

In January 2021, the Phase 2 INVOKE-2 study started enrolling 328 people with early AD to compare monthly infusions of 15, 40, or 60 mg/kg of AL002 to placebo on the CDR-SB as a primary outcome. The treatment phase ran for 48 to 96 weeks, through the end of 2023, at 90 sites in North and South America, Europe, Australia, and New Zealand. In late 2021, Alector reported that amyloid-related imaging abnormalities (ARIA) were observed in INVOKE-2, with some serious neurological adverse events in people with two copies of the ApoE4 allele. The company discontinued dosing E4 homozygotes in the trial, and amended the protocol to exclude them (Feb 2022 press release). After exclusion of homozygotes, no additional serious adverse events related to ARIA were reported (Aug 2022 press release, Jan 2023 company presentation, PDF icon slides 23-32). The company reported more safety data at the July 2023 AAIC in Amsterdam (Aug 2023 conference news). Three serious ARIA-like events in E4 homozygotes included focal neurological deficits, seizures, and mental status changes requiring hospitalization. All resolved with AL002 stoppage and corticosteroid treatment. After E4 homozygotes were excluded from the trial, the overall frequency and severity of ARIA was reduced, although ARIA-E still occurred in 29 percent of participants, and ARIA-H in 27 percent. ARIA occurred more in E4 carriers than non-carriers. It mainly appeared in the first three months of treatment, was asymptomatic, and resolved within 4 months. MRI results from two patients who developed ARIA-H showed signs of microhemorrhage appearing 3 and 5 months after the last AL002 dose. Recruitment for this trial was completed in September 2023. It finished in September 2024. A one-year open label extension was fully enrolled and expected to run through 2025.

On November 25, 2024, Alector announced that INVOKE-2 had failed to meet its primary endpoint of slowing decline on the CDR-SB (press release). Treatment did not improve any secondary endpoints, AD biomarkers, or amyloid PET, despite evidence of sustained target engagement and microglia activation. The company stopped the long-term extension of this trial.

For all trials of AL002, see clinicaltrials.gov.

Last Updated: 26 Nov 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

SHR318

Tools

Back to the Top

Species: Rat
Genes: MAPT
Modification: MAPT: Transgenic
Disease Relevance: Alzheimer's Disease
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

  • Neuronal Loss

No Data

  • Plaques
  • Gliosis
  • Synaptic Loss
  • Changes in LTP/LTD

Plaques

No data.

Tangles

Argyrophilic neurofibrillary tangles are particularly prominent in the brainstem and spinal cord.

Synaptic Loss

No data.

Neuronal Loss

Neuron numbers in the hippocampi and brainstem gigantocellular reticular nucleus do not differ between 10.5-month SHR318 rats and non-transgenic rats.

Gliosis

No data.

Changes in LTP/LTD

No data.

Cognitive Impairment

At 4.5 months, rats show normal learning, but deficits in spatial memory, in the Morris water maze.

Last Updated: 07 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

Subscribe to ALZFORUM RSS