Therapeutics

LU AF82422

Tools

Back to the Top

Overview

Name: LU AF82422
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: alpha-synuclein
Condition(s): Parkinson's Disease, Multiple System Atrophy
U.S. FDA Status: Parkinson's Disease (Phase 1), Multiple System Atrophy (Phase 2)
Company: Genmab A/S, Lundbeck

Background

LU AF82422 is a humanized monoclonal IgG1 antibody targeting the C-terminal of α-synuclein. Genetic and pathology evidence implicate aggregated forms of this protein in the molecular pathogenesis of Parkinson’s disease and other α-synucleinopathies such as dementia with Lewy bodies (DLB).

Preclinical evaluation established safety and CSF target engagement in rats and cynomolgus monkeys. Blood cells express α-synuclein, but the antibody did not bind to most types of human blood cell. It did bind to a small subset of monocytes, but reportedly neither inhibited nor activated them (Fjord-Larsen et al., 2021).

LU AF82422 is one of several α-synuclein antibodies being investigated for PD. Others include ABBV-0805cinpanemabTAK-341prasinezumab, and UCB7853.

Findings

Lundbeck began a Phase 1 single-dose safety study of LU AF82422 in July 2018. The study enrolled 59 healthy Japanese and non-Japanese volunteers and 15 people with Parkinson’s disease at four sites in the U.S. Healthy participants received infusions of one of six doses of antibody ranging from 75 to 9,000 mg, or placebo, followed by 12 weeks of observation. Parkinson’s patients received 2,250 or 9,000 mg, or placebo. The trial also measured exposure and pharmacokinetics of the antibody in blood. It was completed in July 2021, and results were published after peer review (Buur et al., 2024). The treatment was safe and well-tolerated, with no serious adverse events, or adverse events leading to study withdrawal. Pharmacokinetics were dose-proportional and did not differ between the healthy and PD volunteers. The antibody was detected in CSF at 0.1 to 0.5 percent of plasma levels. Two participants developed anti-drug antibodies, which did not affect pharmacokinetics. In a target engagement analysis, the antibody bound to and reduced levels of free α-synuclein in blood and CSF. The PD patients showed a 37 percent reduction in the ratio of free to total synuclein in CSF three weeks after dosing.

In 2021, this antibody received orphan drug designation from the European Medicines Agency.

In November 2021, a Phase 2 study called AMULET began enrolling 64 patients with MSA. Participants were randomized 2:1 to monthly infusions of 4,200 mg Lu AF82422 or placebo for 48 to 72 weeks. The primary outcome is change in the Unified Multiple Systems Atrophy Rating Scale at the end of treatment. Seventeen secondary outcomes span additional measures of symptoms, daily function, global impression, falls, and quality of life, plus volumetric MRI, neurofilament light chain concentrations, and pharmacokinetics. The placebo-controlled part of this trial, running at 19 sites in the U.S. and Japan, finished in November 2023. According to results presented at the March 2024 AD/PD conference, the trial showed a trend on the primary endpoint but, at 19 percent slowing of decline, missed statistical significance. There were trends to slowing on secondary endpoints related to functional and motor deterioration. A 27 percent slowing of progression on the FDA-preferred endpoint of the modified USMARS just missed statistical significance. A post hoc subgroup analysis identified a statistically significant 42 percent slowing on the modified UMSARS in the less impaired subgroup. Other trends were slowing of brain volume loss in the pons and cerebellum, and a greater lowering of NfL in the CSF. Of 61 patients who began therapy, 49 completed the full 72 weeks. No safety concerns were identified, and no antidrug antibodies were elicited. A one-year, open-label extension is ongoing. Lundbeck intends to run a Phase 3 study (company release).

In February 2024, Lundbeck registered an additional Phase 1 trial to assess safety, tolerability, pharmacokinetics and immunogenicity of a single infusion of Lu AF82422 in healthy Caucasian and Chinese adults. It will run from March to September 2024. 

For details on LU AF82422 trials, see clinicaltrials.gov.

Last Updated: 03 May 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Suvorexant

Tools

Back to the Top

Overview

Name: Suvorexant
Synonyms: Belsomra, MK-4305
Chemical Name: [(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl]methanone
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Approved)
Company: Merck
Approved for: Insomnia, Insomnia in mild to moderate Alzheimer's disease

Background

This orexin receptor antagonist was approved by the U.S. FDA in 2014 to treat insomnia. In February 2020, Suvorexant became the first medication to be approved for treating sleep disorders in Alzheimer’s disease (company press releaseFDA prescribing information). Suvorexant is taken by mouth.

Suvorexant was reported to have a lower potential for abuse, and be generally safer, than other sleep medications frequently used in aging populations, such as benzodiazepines (Okino et al., 2023Moline et al., 2023Pan et al., 2023Katsuta et al., 2023).

Orexin is a neuropeptide produced by the hypothalamus. It promotes wakefulness, and blocking its receptor promotes sleep. 

Many people with AD suffer from disruption of circadian rhythms that leads to poor sleep, nighttime activity and daytime sleepiness. Circadian disruption is a frequent cause of institutionalization (Harper et al., 2005). Changes in sleep/wake cycles also occur in preclinical AD, and are linked to increased amyloid deposition and risk of cognitive decline (Oct 2013 news; Ju et al., 2013; Musiek et al., 2018). Thus, sleep aids are of interest as potential disease modifiers. CSF orexin levels are elevated in Alzheimer's disease (Oct 2014 news).

In the APP/PS1 mouse model of AD, suvorexant was shown to improve circadian rhythms and cognitive function, restore hippocampal synaptic plasticity, and reduce Aβ plaque deposition in hippocampus and cortex (Zhou et al., 2020).

Findings

In May 2016, Merck began a Phase 3 trial to test whether suvorexant would ease sleep disturbances in 285 people with mild to moderate AD and insomnia. Participants took placebo for two weeks, followed by a baseline overnight polysomnography sleep study. They were then randomized to suvorexant or placebo for four weeks, starting at 10 mg daily and escalating after two weeks to 20 mg as tolerated. After four weeks, participants underwent a final overnight polysomnography session.

In May 2019, company scientists presented results at the American Academy of Neurology meeting (May 2019 conference news). Suvorexant was efficacious on both primary outcomes. It lengthened total nighttime sleep time by 28 minutes compared with placebo, and shortened nighttime waking time by 15 minutes. Importantly, suvorexant did not impair cognition. Adverse events occurred in fewer than 5 percent of participants, and included daytime drowsiness, dry mouth, and falls. Trial data are published (Herring et al., 2020).

In late 2016, a study conducted at Washington University, St. Louis, started to measure whether improving sleep efficiency with suvorexant affects CSF Aβ. Thirty-eight cognitively normal participants took 10 or 20 mg suvorexant or placebo on two consecutive nights, and slept in a research hospital while undergoing repeated CSF sampling to monitor Aβ production, clearance, and concentration, and tau levels. The study was completed in March 2021. According to published results, 20 mg suvorexant decreased the phospho-tau181/tau ratio by 10-15 percent within hours, but did not affect p-tau202 or p-tau217 to tau ratios. The drug lowered Aβ by 10 to 20 percent compared to placebo starting five hours after dosing (Lucey et al., 2023).

In May 2022, a Phase 2 study began at Washington University to assess the effect of two years of suvorexant on the accumulation of brain amyloid. The study plans to enroll 200 cognitively normal participants with PET evidence of amyloid accumulation and poor sleep, who will take 20 mg suvorexant or placebo nightly. The primary outcome will be change in brain amyloid by PET scan. Secondary outcomes include tau PET, cognitive measures, and changes in plasma and CSF markers of Aβ and tau, as well as blood and CSF transcriptomic, proteomic, and lipidomic analysis, and microbiome evaluation. Completion is anticipated in May 2026.

Suvorexant is also being evaluated for insomnia related to Parkinson’s disease, progressive supranuclear palsy, opioid and alcohol use disorder, multiple sclerosis, PTSD, major depressive disorder, and restless legs syndrome.

For details on suvorexant trials, see clinicaltrials.gov.

Last Updated: 15 May 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Bifidobacterium breve A1

Tools

Back to the Top

Overview

Name: Bifidobacterium breve A1
Synonyms: MCC1274, B. breve
Therapy Type: Supplement, Dietary (timeline)
Target Type: Other (timeline)
Condition(s): Mild Cognitive Impairment
U.S. FDA Status: Mild Cognitive Impairment (Phase 2)
Company: Morinaga Milk Industry Co., Ltd

Background

Bifidobacterium breve A1 is a proprietary strain of probiotic bacteria isolated from the intestinal tract of an infant. A main component of the human gut microbiome, Bifidobacteria species form part of  many probiotic supplements widely used to treat gastrointestinal and other ailments (see WebMD). Bifidobacteria are associated with anti-inflammatory and immune-modulatory effects that are generally thought to promote health, but are not understood in detail.

Some studies have reported that intestinal flora influence microglia activity, brain amyloid deposition, and cognition in mouse models of Alzheimer’s disease and in people, implying a potential for probiotic use in preventing dementia (Apr 2020 conference news; Cattaneo et al., 2017).

In preclinical work, feeding mice Bifidobacterium breve A1 prevented cognitive deficits after brain injection of Aβ peptide. Probiotic treatment suppressed Aβ-mediated induction of inflammatory and immune genes in the hippocampus.  (Kobayashi et al., 2017).

Several different probiotic mixtures containing Bifidobacterium yielded broadly similar results in rodent models (Mehrabadi and Sadr, 2020Wang et al., 2020Yang et al., 2020Rezaeiasl et al. 2019).

The mechanisms by which probiotic supplementation affects the brain remains to be clarified. In Kobayashi et al., 2017, the effects of B. breve were partially mimicked by giving mice non-viable bacteria, or the bacterial metabolic product acetate.

Findings

A double-blind trial, conducted in 38 cognitively normal Japanese people in their 60s and 70s, evaluated whether a 12-week regimen of probiotic Bifidobacteria supplementation combined with resistance training exercise improved cognitive functions and other health outcomes. The combined intervention was reported to have led to improvement on the Japanese version of the MoCA screen, as well as lower depression/anxiety scores and body mass index, over placebo (Inoue et al., 2018). 

In an open-label pilot study in Japan, 27 elderly people with mild cognitive impairment took capsules of B. breve A1 daily for 24 weeks. Nineteen completed the study. Their Mini-Mental State Exam (MMSE) scores improved, as did measures of mood and gastrointestinal function (Kobayashi et al., 2019).

A subsequent double-blind trial enrolled 121 elderly Japanese with self-reported memory complaints and MMSE scores between 22 and 27. They ingested two capsules per day of freeze-dried B. breve, containing a total of 2 x 1010 viable bacteria, or placebo, for 12 weeks. Dual endpoints were change in the Japanese version of the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and MMSE. The treatment had no effect overall, as the placebo and B. breve groups improved equally on both measures. When the investigators analyzed subgroups with high RBANS (above 41, in the cognitively normal range), or low RBANS (below 41, in the MCI range) scores at baseline,  the latter showed improvement on the MMSE compared with placebo (Kobayashi et al., 2019).

A second placebo-controlled study in Japan enrolled 80 people age 50 to 79 with suspected mild cognitive impairment and RBANS scores between 11 and 45. Participants received the same daily B. breve or placebo regimen as the previous trial, for 16 weeks. The primary endpoint was change in Japanese RBANS; the Japanese version of the MCI screen (JMCIS) was a secondary measure. All but one person completed the study, with no adverse events reported. Treatment resulted in a 17.6-point improvement in RBANS, compared with a 5.9-point improvement in the placebo group. Treatment was associated with score increases over placebo in immediate memory, visuospatial/constructional, and delayed memory subscales. The JMCIS showed a trend toward more improvement in treated versus placebo (Xiao et al., 2020).

No completed or ongoing B. brevis A1 trials are found in registries. Other Bifidobacterium strains are being evaluated in registered trials for obesity and other metabolic disorders, gastrointestinal conditions, and autism. For more details, see clinicaltrials.gov.

Last Updated: 16 Jul 2020

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

PS19 with humanized TREM2 (R47H)

Synonyms: PS19-T2R47H, PS19-TREM2R47H

Tools

Back to the Top

Species: Mouse
Genes: MAPT, TREM2, Trem2
Modification: MAPT: Transgenic; TREM2: Transgenic; Trem2: Knock-Out
Disease Relevance: Alzheimer's Disease, 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

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

Plaques

No data.

Tangles

Tangles revealed using antibody PG5 at 9 months.

Synaptic Loss

At 9 months of age, more synapses and fewer dystrophic synapses, compared with PS19 mice carrying the common variant of TREM2, but no data relative to wild-type mice.

Neuronal Loss

No data relative to wild-type mice, but at 9 months of age, the volumes of the hippocampus and entorhinal/piriform cortex are larger, and the granule cell layer of the dentate gyrus and pyramidal cell layer of the piriform cortex are thicker, in PS19-TREM2R47H mice, compared with PS19 mice carrying the common variant of human TREM2.

Gliosis

At 9 months of age, decreased expression of markers of astroglial and microglial reactivity, compared with PS19 mice carrying the common variant of TREM2, but no data relative to wild-type mice.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 14 Jul 2020

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

PS19 with humanized TREM2 (common variant)

Synonyms: PS19-TREM2CV, PS19-T2CV

Tools

Back to the Top

Species: Mouse
Genes: MAPT, TREM2, Trem2
Modification: MAPT: Transgenic; TREM2: Transgenic; Trem2: Knock-Out
Disease Relevance: Alzheimer's Disease, 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

  • Plaques
  • Changes in LTP/LTD
  • Cognitive Impairment

Plaques

No data.

Tangles

Tangles revealed using antibody PG5 at 9 months.

Synaptic Loss

Fewer synapses and more dystrophic synapses, compared with PS19 mice carrying the R47H variant of TREM2.

Neuronal Loss

At 9 months, atrophy of hippocampus and entorhinal/piriform cortex and pronounced ventricular expansion. Thinning of the granule cell layer of the dentate gyrus and pyramidal cell layer of the piriform cortex, compared with PS19 mice carrying TREM2-R47H.

Gliosis

Elevated expression of markers of astroglial and microglial reactivity, compared with PS19 mice carrying the R47H variant of TREM2.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 14 Jul 2020

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, humanized (R47H)

Synonyms: R47H+mTrem2−/−, R47H-KO, TREM2R47H

Tools

Back to the Top

Species: Mouse
Genes: TREM2, Trem2
Modification: TREM2: Transgenic; Trem2: Knock-Out
Disease Relevance: Alzheimer's Disease
Strain Name: N/A

Summary

People who carry one copy of the R47H variant of TREM2 have an approximately threefold greater risk of developing Alzheimer’s disease than do people homozygous for the common variant. This mouse model, referred to here as TREM2R47H, expresses the R47H variant of human TREM2 in the absence of mouse Trem2. This line has been crossed with models of amyloidosis and tauopathy (see Related Strains below).

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

  • Gliosis

No Data

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

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Neuronal Loss

No data.

Gliosis

Expression of DAM (disease-associated microglia) genes is low at 8.5 months, suggesting that microglia are in a resting or homeostatic state.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 14 Jul 2020

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, humanized (common variant)

Synonyms: CV+mTrem2−/−, CV-KO, TREM2CV

Tools

Back to the Top

Species: Mouse
Genes: TREM2, Trem2
Modification: TREM2: Transgenic; Trem2: Knock-Out
Disease Relevance: Alzheimer's Disease
Strain Name: N/A

Summary

This mouse model, referred to here as TREM2CV, expresses the common variant of human TREM2 in the absence of mouse Trem2. Transgenic TREM2 is expressed in microglia and macrophages, where it was shown to function properly in an ex vivo assay. This line has been crossed with models of amyloidosis and tauopathy (see Related Strains below).

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

  • Gliosis

No Data

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

Plaques

No data.

Tangles

No data.

Synaptic Loss

No data.

Neuronal Loss

No data.

Gliosis

Expression of DAM (disease-associated microglia) genes is low at 8.5 months, suggesting that microglia are in a resting or homeostatic state.

Changes in LTP/LTD

No data.

Cognitive Impairment

No data.

Last Updated: 14 Jul 2020

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

QALSODY™

Tools

Back to the Top

Overview

Name: QALSODY™
Synonyms: Tofersen, BIIB067, Ionis-SOD1Rx , ASO1
Therapy Type: DNA/RNA-based
Target Type: Other (timeline)
Condition(s): Amyotrophic Lateral Sclerosis
U.S. FDA Status: Amyotrophic Lateral Sclerosis (Approved)
Company: Biogen, IONIS Pharmaceuticals
Approved for: ALS

Background

This drug is a second-generation antisense oligonucleotide (ASO) targeting the mRNA for superoxide dismutase 1. It mediates mRNA degradation to prevent SOD1 protein synthesis and reduce levels of SOD1 protein. Tofersen is being developed for ALS caused by SOD1 mutations, which account for about 20 percent of all familial ALS and 2 percent of all ALS cases. Although the exact pathological mechanism remains unknown, mutant SOD1 is believed to exert a toxic action on motor neurons, and reducing its levels may be beneficial.

In 2013, Ionis tested a first-generation SOD1 ASO, which proved safe in people, but lacked potency (May 2013 news). The company then screened 2,000 SOD1 ASOs and identified two targeting the mRNA 3' untranslated region that were most effective at lowering SOD1 mRNA and protein levels in cells. Injection of ASO1 into the brains or spinal cords of mice or rats expressing mutant SOD1 reduced spinal-cord levels of SOD1 mRNA and protein. A single injection delayed disease onset, improved motor function, and increased survival in the animals. ASO1 treatment preserved neuromuscular innervation and stemmed the rise of phosphorylated neurofilament heavy chain in blood, a prognostic biomarker for ALS. In nonhuman primates, ASO1 lowered SOD mRNA in the CNS after spinal-cord injection (Jul 2018 news on McCampbell et al., 2018).

Biogen licensed ASO1 from Ionis, and is leading its clinical development.

Findings

In January 2016, Biogen and Ionis began a Phase 1/2 safety trial in SOD1-ALS. The study enrolled 84 people who received single- or multiple-ascending doses by intrathecal injection into their lumbar spinal cord. In the multiple-dosing phase, and 48 participants in four cohorts were randomized 3:1 to receive 20, 40, 60, or 100 mg BIIB067 or placebo five times over 12 weeks. Primary outcomes were adverse events and PK; the secondary outcome was levels of SOD1 protein in cerebrospinal fluid after the last dose. The study took place at 18 sites in the U.S., Canada, and Western Europe. 

The multiple-dosing phase of the trial was completed in January 2019. According to results presented at the May 2019 American Academy of Neurology conference, and later published, ASO treatment was safe and reduced mutant SOD1 protein (May 2019 conference news; Miller et al., 2020). Most adverse events were mild to moderate; the most common were headache, procedural pain, and postlumbar syndrome related to the spinal taps. Falls were also common. Seven participants had serious adverse events. Three people died. One person in the treatment group and one on placebo died of respiratory failure; one person in the treatment group died from a lung embolism. SOD1 protein levels in cerebrospinal fluid dropped by 3 percent in the low-dose group, and 36 percent in the high-dose group, compared with placebo.

Approximately 10 percent of people receiving BIIB067 showed elevated white cell counts in their CSF, and a similar number had elevated protein in CSF. No one developed myelitis, an inflammation of the spinal cord accompanied by sensory and motor deficits, but the investigators noted this condition was seen in some people dosed with BIIB067 in an ongoing Phase 3 and long-term extension (see below).

The small trial was not designed to measure efficacy, but exploratory outcomes included change from baseline in the ALS Functional Rating Scale-Revised (ALSFRS-R), lung vital capacity and muscle strength, and blood and CSF neurofilament concentrations. The highest-dose group appeared to decline more slowly on the ALSFRS-R, showing a loss of 1.19 points at week 12, compared with 5.63 in the placebo group. Concentrations of neurofilament heavy and light chains in CSF and blood decreased from baseline to 12 weeks in the highest-dose group.

In March 2019, Biogen added a Phase 3 efficacy arm to the same trial. Called VALOR, the study enrolled 108 people with SOD1-ALS, of whom 72 received eight infusions of 100 mg BIIB067 over 28 weeks; 36 received placebo. The primary outcome was change from baseline in the ALSFRS-R at week 28 in a subgroup of patients defined as faster progressing using trial-specific prognostic criteria. Secondary outcomes were other clinical measures such as time to needing a ventilator, muscle strength, survival, CSF SOD1, and plasma phosphorylated neurofilament heavy chain. The trial was conducted at 32 sites in North America, Europe, Australia, and Japan, and finished in July 2021. An ongoing, open-label safety extension involving 139 patients will run through August 2024, with treatment offered for up to five years.

In December 2018, a research study began to evaluate the distribution of BIIB067 in the central nervous system by SPECT/CT imaging after co-administration of a microdose of radiolabeled BIIB067 and high- or low-dose BIIB067 in 20 healthy volunteers. Recruitment was suspended in May 2020, due to the COVID-19 pandemic, and the study was completed in July 2021 with just eight participants.

In May 2021, recruitment began for a Phase 3 study in presymptomatic carriers of an SOD1 mutation. Called ATLAS, this study intends to enroll 150 people, who must also have elevated plasma NfL. After a run-in period to confirm NfL elevation, participants will receive 100 mg tofersen or placebo via intrathecal injection three times in the first month, and then monthly for up to two years. The primary endpoint is the number of participants who develop ALS symptoms within the first year of treatment. Secondary outcomes include the time to emergence of symptoms, ALSFRS-R, lung function, need for ventilation, death, adverse events, and changes in SOD1 and neurofilament light chain (NfL). Participants who become symptomatic during the trial will be eligible for open-label treatment for up to two years. The study will run through August 2027. The protocol is published (Benatar et al., 2022).

On October 17, 2021, Biogen announced that the VALOR trial had missed its primary endpoint of slowing decline on the ALSFRS-R at week 28 (Oct 2021 news). Full trial data was subsequently published (Miller et al., 2022). The efficacy analysis was based on 60 fast progressors, with 39 receiving tofersen, 21 placebo. Treatment was associated with a numerical improvement of ALSFRS and other endpoints, but it fell short of statistical significance. Tofersen reduced mutant CSF SOD1 by 29 percent, compared to a 16 percent increase with placebo. Plasma NfL declined by 60 percent in the treated group, and increased by 20 percent in the placebo group. The groups were unbalanced at baseline, with the treated group starting with about 15 percent higher NfL than those who got placebo. In an open-label extension, patients who switched from placebo to treatment had, by the 12-month time point, decreases in CSF SOD1, NfL, and a slowing in their rate of decline on multiple clinical measures.

In the trial, adverse events were mostly related to spinal infusions or ALS, except that more tofersen recipients reported arm, leg, or back pain. Tofersen caused serious neurological side effects in 7 percent of people, including inflammation around nerves or the spinal cord, nerve pain, and increased intracranial pressure (e.g., see Reilich et al., 2024). More than half of tofersen recipients had at least one instance of elevated white cells in their CSF. Median CSF protein concentration increased in the treated, but not placebo, participants. Separate case reports identified macrophages with numerous cytoplasmic inclusions accounting for more than half of the CSF infiltrating cells, similar to cells previously seen during treatment with the ASO nusinersen in people with spinal muscular atrophy (Sparasci et al., 2023, Vidovic et al., 2024).

On July 26, 2022, Biogen and Ionis announced that the FDA had accepted a new drug application for tofersen, granting it priority review. Biogen sought accelerated approval based on changes in the surrogate biomarker NfL (press release). The FDA set a decision date of January 25, 2023.

On April 26, 2023, the agency granted tofersen accelerated approval for use in people with SOD1-ALS, after an advisory committee unanimously agreed that the drop in NfL in VALOR was likely predictive of a clinical benefit (April 2023 news). Per FDA rules, the company is obliged to show efficacy by way of the ongoing Phase 3 Atlas trial, which will run until 2027. The new drug will be marketed under the name QALSODY™.

In May 2024, QALSODY™ was approved for sale in the European Union (press release).

Several studies are following the experience of patients receiving tofersen in the clinic. In one cohort of 17 patients who were treated for at least a year, progression slowed significantly during treatment compared to before (Sabatelli et al., 2024). Nine stabilized or slightly improved on the ALSFLS-R, while four deteriorated. The rest progressed too slowly to detect significant changes. As in the VALOR trial, most patients markedly reduced their serum NfL. Just over half had increased levels of white blood cells and protein in CSF, and two developed symptomatic nerve inflammation that responded to steroids. A follow-up of German patients confirmed a decline in serum NfL with clinical use of tofersen (Wiesenfarth et al., 2024; Meyer et al., 2023). NfL reductions occurred in patients who were homozygous or heterozygous for the D91A SOD1 mutation, suggesting a causative role for the mutation even in heterozygotes (Weishaupt et al., 2024). Improvements in clinical and patient-reported outcomes led to high patient satisfaction and recommendation of tofersen (Meyer et al., 2024). Tofersen treatment increased levels of CSF selenium, potentially indicating improved CNS redox status (Vinceti et al., 2024).

For details on BIIB067 trials, see clinicaltrials.gov.

Last Updated: 02 Aug 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Lu AF87908

Tools

Back to the Top

Overview

Name: Lu AF87908
Synonyms: hC10.2
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Tau (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 1)
Company: Lundbeck

Background

Lu AF87908 is a humanized mouse IgG1 monoclonal antibody to phosphorylated tau protein. It was generated by immunization of mice with a tau peptide spanning residues 386-408 and phosphorylated at serine 396 and 404.

In preclinical work, the mouse version of this antibody, C10.2, preferentially bound hyperphosphorylated tau aggregates from brain, and reduced the ability of brain-derived tau to seed aggregation in cultured neurons and in rTg4510 tau transgenic mice (Rosenqvist et al., 2018). The antibody mediated uptake and lysosomal degradation of mouse-brain-derived pathological tau aggregates in primary microglia in culture; notably, this required effector function via the antibody's Fcγ receptor (Andersson et al., 2019). The humanized antibody bound to phosphorylated tau in postmortem brain from cases of AD and primary tauopathies, and prevented seeding of aggregation by brain extracts (Helboe et al., 2022). Intravenous administration caused a dose-dependent decrease in CSF pS396-tau in rTg4510 mice (Jacobsen et al., 2023).

Findings

In September 2019, Lundbeck initiated a Phase 1 single-dose study of Lu AF87908. Conducted at six sites in the U.S., the placebo-controlled trial plans to enroll 86 adults in three sequential cohorts: healthy, healthy Japanese and Chinese, and patients with Alzheimer’s disease. Safety and plasma antibody concentrations will be monitored for three months after infusion. The study is scheduled to run through June 2023.

For details on Lu AF87908 trials, see clinicaltrials.gov.

Last Updated: 28 Feb 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

PNT001

Tools

Back to the Top

Overview

Name: PNT001
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Tau (timeline)
Condition(s): Alzheimer's Disease, Traumatic Brain Injury
U.S. FDA Status: Alzheimer's Disease (Inactive), Traumatic Brain Injury (Inactive)
Company: Pinteon Therapeutics

Background

PNT001 is a monoclonal antibody to the cis isomer of tau phosphorylated at threonine 231, a form of pathological tau suspected to play a role in traumatic brain injury, chronic traumatic encephalopathy, vascular dementia, and Alzheimer’s disease.

Phosphorylated T231 tau exists in cis and trans conformations, as modulated by the peptidyl-prolyl cis/trans isomerase Pin1 (Galas et al., 2006Hamdane et al, 2006). Cis pT231 has been detected in brain tissue from people with AD and CTE and increases acutely after traumatic brain injury in humans and rodent models. In humans, its levels in CSF correlate with severity of brain injury (Sisakht et al., 2022). Cis pT231 is resistant to dephosphorylation and degradation, and promotes aggregation (Nakamura et al., 2012; reviewed in Lu et al., 2016). 

In preclinical models of brain injury, a mouse monoclonal antibody to cis pT231 prevented axonal pathology, astrogliosis, tau oligomerization and tangle formation, brain atrophy, and behavioral and other deficits (Kondo et al., 2015; Albayram et al., 2017; Albayram et al., 2019).

Cis pTau appears to contribute to vascular dementia. It was found to be elevated in brains of people with vascular dementia, who lacked tau tangles. In mouse models of vascular dementia, a cis-tau targeted monoclonal antibody reduced neurodegeneration and improved cognitive impairment. The cis mAb ameliorated progression of AD-like neurodegeneration and cognitive impairment in mice expressing human tau (Qui et al., 2021).

Findings

In September 2019, Pinteon began a Phase 1 safety and tolerability study of PNT001 in healthy adults. In the trial, 49 people received single doses of 33, 100, 300, 900, 2,700, or 4,000 mg antibody or placebo, infused over 30 to 60 minutes. Primary outcomes were adverse events and abnormalities on clinical and laboratory measures after 16 weeks. Other outcomes included antibody concentration in serum and CSF, measures of total tau, cis pT231 tau, pT231 tau, and NfL in CSF, as well as serum NfL and antidrug antibodies. According to data presented at AD/PD 2021, the antibody produced dose-linear blood and CSF concentrations that stayed constant for 28 days, and was well tolerated. For doses of 900 mg and above, CSF antibody concentrations exceeded the minimum required for tau binding (Mar 2021 conference news).

In March 2021, the company began a Phase 1 study of multiple ascending doses in 64 hospitalized patients with acute traumatic brain injury. Each patient was to receive three doses of 1,000 or 4,000 mg, or placebo. The first dose was planned to occur within 24 hours of injury; the time frame for subsequent doses was not specified. Safety, tolerability, pharmacokinetic, biomarker, imaging and cognitive data were to be collected over 12 weeks. The trial was terminated for non-safety reasons shortly after it began, and enrolled only one patient.

For details on PNT001 trials, see clinicaltrials.gov.

Last Updated: 28 Jun 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Subscribe to ALZFORUM RSS