Research Models

hAβ-KI

Synonyms: hAbeta-loxP-KI

Tools

Back to the Top

Species: Mouse
Genes: App
Modification: App: Knock-In
Disease Relevance: Alzheimer's Disease
Strain Name: B6(SJL)-Apptm1.1Aduci/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

  • Plaques
  • Neuronal Loss
  • Gliosis

No Data

  • Tangles

Plaques

No plaques observed through 22 months of age, using immunohistochemical, thioflavin-S or Congo red stains.

Tangles

No data.

Synaptic Loss

Fewer synaptophysin-immunoreactive puncta, but similar numbers of PSD95-immunoreactive puncta, in knock-in mice compared with wild-type mice.

Neuronal Loss

Neuron numbers in hippocampal CA1 were similar in 22-month hAβ-KI and wild-type mice, although hippocampal volume was decreased in the knock-in mice.

Gliosis

Neither microgliosis nor astrogliosis was observed through 22 months of age.

Changes in LTP/LTD

Impaired theta-burst-induced LTP at Schaffer collateral-CA1 synapses, by 18 months of age.

Cognitive Impairment

Differed from wild-type mice in the contextual fear conditioning test by 10 months of age and in the novel object recognition task by 14 months.

Last Updated: 11 Jun 2021

Further Reading

No Available Further Reading

Therapeutics

UCB7853

Tools

Back to the Top

Overview

Name: UCB7853
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: alpha-synuclein
Condition(s): Parkinson's Disease
U.S. FDA Status: Parkinson's Disease (Phase 1)
Company: UCB S.A.

Background

UCB7853 is an α-synuclein antibody. No further information is available on its origin or specificity.

Findings

In December 2020, UCB began a Phase 1 study in healthy men and people with Parkinson's. The study plans to enroll 64 participants in the U.K., and to evaluate single- and multiple-ascending doses of UCB7853 infusion for safety, tolerability, and pharmacokinetics. In June 2021, UCB suspended enrollment for further evaluation, and changed the trial's anticipated completion from September 2022 to April 2023.

For details, see clinicaltrials.gov.

Last Updated: 20 Jul 2021

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

BIIB078

Tools

Back to the Top

Overview

Name: BIIB078
Synonyms: BIIB-078, IONIS-C9Rx
Therapy Type: DNA/RNA-based
Target Type: Other (timeline)
Condition(s): Amyotrophic Lateral Sclerosis
U.S. FDA Status: Amyotrophic Lateral Sclerosis (Discontinued)
Company: Biogen, IONIS Pharmaceuticals

Background

BIIB078 is an antisense oligonucleotide targeting the chromosome 9 open reading frame 72 (C9ORF72) gene mRNA. It is being developed for ALS caused by hexanucleotide repeat expansions in C9ORF72 (van Blitterswijk et al, 2012). These expansions are the most common genetic cause of ALS, accounting for about 34 percent of all familial ALS and 12 percent of all ALS cases. The ASO mediates degradation of expansion-containing mRNAs. Both C9ORF72 RNA and the dipeptide proteins produced from the hexanucleotide repeats are believed to be toxic to motor neurons, hence the rationale is that reducing them will be beneficial.

In preclinical work with patient-derived cell lines, C9ORF72 ASOs mitigated RNA toxicity (Donnelly et al., 2013; Sareen et al., 2013; Lagier-Tourenne et al., 2013). In mice expressing the C9ORF72 expansion, a single, intraventricular dose of an ASO targeting the hexanucleotide repeats reduced toxic RNA and dipeptide aggregates, and attenuated behavioral and cognitive deficits (Jiang et al., 2016).

Biogen is developing BIIB078 in collaboration with Ionis Pharmaceuticals.

Findings

In September 2018, Biogen began a first-in-human Phase 1 in adults who have ALS with C90ORF72 expansions. Participants had to have a slow vital capacity above 50 percent of predicted normal and could use riluzole or edaravone, both approved for ALS. The study enrolled 106 participants in six ascending-dose cohorts and placebo, delivered by intrathecal injection five times over an eight-month span. The primary endpoints were adverse events; secondary outcomes included pharmacokinetics, clinical measures of function and muscle strength, and CSF levels of dipeptide proteins and phosphorylated neurofilament heavy chain, a biomarker of motor neuron death. An open-label safety extension offered monthly dosing for two years.

The placebo-controlled portion of the trial ended in November 2021. In March 2022, the companies announced top-line results, according to which the drug was well-tolerated but showed no difference from placebo on clinical endpoints at doses up to 60 mg. The 90 mg dose group trended toward greater decline across secondary endpoints. Based on these results, the companies ended the program, including the ongoing open-label extension (press release). Trial results were published after peer review (van den Berg et al., 2024).

For details on BIIB078 trials, see clinicaltrials.gov.

Last Updated: 10 Sep 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Minzasolmin

Tools

Back to the Top

Overview

Name: Minzasolmin
Synonyms: UCB0599, NPT200-11
Chemical Name: N-[(2R)-1-(1H-indol-3-yl)hexan-2-yl]-2-(4-methylpiperazin-1-yl)-1,3-thiazole-5-carboxamide
Therapy Type: Small Molecule (timeline)
Target Type: alpha-synuclein
Condition(s): Parkinson's Disease
U.S. FDA Status: Parkinson's Disease (Phase 2)
Company: Neuropore Therapies, Inc., Novartis Pharmaceuticals Corporation, UCB S.A.

Background

Minzasolmin is a small-molecule α-synuclein aggregation inhibitor. Oligomerization and aggregation of α-synuclein is implicated in Parkinson’s disease, dementia with Lewy bodies, and multiple-system atrophy. Minzasolmin was discovered at Neuropore, licensed to UCB in 2014, and licensed for co-development to Novartis in 2021.

Neuropore scientists and collaborators have described a series of cyclic peptidomimetic compounds designed to interact with the C-terminal domain of α-synuclein and prevent it from binding to membranes and oligomerizing there. One compound was reported to reduce α-synuclein aggregation in vitro and in α-synuclein transgenic mouse models, where it also normalized neural and inflammatory markers, and ameliorated motor deficits. In vivo imaging showed a reduction in cortical synaptic α-synuclein within one hour of dosing in the mice (Wrasidlo et al., 2016).

Minzasolmin is a second-generation compound, optimized for oral bioavailability and brain entry. It is the purified R enantiomer of the racemic mixture NPT-200-11. In preclinical work, NPT-200-11 reduced retinal α-synuclein pathology over two months of daily administration in mice expressing human α-synuclein. It also lessened cortical α-synuclein pathology and astrogliosis, normalized striatal dopamine transporter levels, and improved motor function (Price et al., 2018). Similar results were subsequently reported for minzasolmin (Price et al., 2023). A high-resolution structure of oligomeric, membrane bound α-synuclein suggests that minzasolmin interacts with the protein to increase its flexibility and impair its embedding into the membrane. In this way, minzasolmin interferes with fibril growth and toxic pore formation, and promotes the release of synuclein monomers in their soluble, random coil form (Schwarz et al., 2023). This study showed that the enantiomers interacted identically with membrane-bound α-synuclein.

Findings

In April 2021, UCB presented the results of a Phase 1b study at the American Academy of Neurology Annual Meeting (Genius et al., 2021). According to a news report, the study enrolled 31 people with early stage Parkinson’s disease, who received 90 or 180 mg UCB0599 or placebo twice daily for 28 days. Most adverse events were mild to moderate, occurring at similar frequency in treated and placebo groups. Two participants treated with UCB0599 showed mild hypersensitivity reactions. Overall, the drug raised no safety concerns, according to the investigators. The company subsequently published this data, along with results of single and multiple ascending doses in 73 healthy volunteers (Smit et al., 2022). At 90, 180, 360, and 450 mg, pharmacokinetics were linear and predictable, including in CSF. Food effects were minimal. Co-administration of the cytochrome P450 inhibitor itraconazole doubled plasma exposure, but did not change short term safety. Two volunteers in this study had moderately severe hypersensitivity reactions.

In December 2020, UCB began an 18-month, Phase 2, placebo-controlled study in 300 people with early PD and mild symptoms. In late 2021, enrollment was increased to 450, split evenly among high-dose, low-dose, and placebo arms, delivered as granules in capsules.. The primary outcome is change in the Unified Parkinson’s Disease Rating Scale (UPDRS), Parts I-III. Secondary outcomes include UPDRS individual parts I, II, and III, time to disease worsening, change in Montreal cognitive assessment, dopamine transporter imaging, time to start of symptomatic treatment, symptomatic treatment intake, adverse events, and events leading to withdrawal. The trial enrolled 496 participants at more than 100 sites in the U.S., Canada, France, Germany, Italy, Poland, Spain, the Netherlands and the U.K., and was completed in September 2024. A long-term extension has enrolled 428 participants, at either high or low dose.

A Phase 1 study evaluated 11[C]-labeled UCB0599 as a PET ligand. Four participants were scanned before, and two hours after, a 360 mg oral dose of unlabeled UCB0599. The radioligand distributed throughout the brain, and steady-state brain concentrations were approximately half of plasma levels. Prior administration of unlabeled compound did not change the amount of tracer entering the brain (Mercier et al., 2023).

In May 2023, a Phase 1 began to compare the bioavailability of two new formulations of UCB0599 under normal and elevated gastric pH. A second part of the study assesses pharmacokinetics in healthy Japanese and Chinese participants. This study was completed in April 2024.

In July 2024, the company registered a Phase 1 study to assess the bioavailability of a new tablet formulation, and the effect of food. The trial plans to enroll 18 healthy adults, and run until October 2024.

For details on minzasolmin/UCB0599 trials, see clinicaltrials.gov.

Last Updated: 16 Oct 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Edaravone

Tools

Back to the Top

Overview

Name: Edaravone
Synonyms: MCI-186, Radicava®, Radicut®, MT-1186, FAB122
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Amyotrophic Lateral Sclerosis
U.S. FDA Status: Amyotrophic Lateral Sclerosis (Phase 4)
Company: Mitsubishi Tanabe Pharma
Approved for: Amyotrophic Lateral Sclerosis

Background

Edaravone is an antioxidant drug marketed to treat amyotrophic lateral sclerosis. A pyrazolone free-radical scavenger, it reduces oxidative damage, which is believed to contribute to neuron death in ALS.

Edaravone was approved in Japan in 2015, and in the U.S. in 2017, based primarily on results of a single clinical trial in early stage ALS patients conducted in Japan (May 2017 news; FDA review documentCruz, 2018). The European Medicines Agency declined to approve edaravone, citing a lack of confidence that the Japanese data would translate into a clinical benefit to the broader European patient population (EMA review document). The drug previously had been approved in 2001 in Japan for acute treatment of neurological symptoms and functional impairment due to ischemic stroke.

For ALS, the drug is given in cycles of intravenous infusions daily for two weeks, followed by a two-week break. It can cause hives, swelling, and shortness of breath in some people, due to anaphylactic reactions to sulfite-containing infusion components. Otherwise, it is well tolerated. Common side effects are bruising, problems walking, and headaches.

In May 2022, the FDA approved an oral formulation of this drug, Radicava ORS; edavarone is not approved by the European Medicines Agency (Dorst and Genge, 2022).

In ALS animal models, edaravone inhibits motor neuron death by reducing oxidative stress (Ito et al., 2008).

Oxidative stress plays a role in Alzheimer’s disease, as well, and edaravone has shown beneficial effects in multiple cellular and animal models of Aβ toxicity (e.g., Feng et al., 2019Feng et al., 2020Jiao et al., 2015).

Findings

For the first trial in ALS, Mitsubishi Tanabe conducted an open-label, Phase 2 study in 20 patients. Participants received six cycles of 30 or 60 mg edaravone by daily infusion for two weeks, alternating with a two-week pause, over six months. The primary endpoint was change in the ALS Functional Rating Scale (ALSFRS-R) score over six months of treatment, compared to the six months before treatment. According to published results, 60 mg edaravone significantly slowed the decline in ALSFRS scores, compared to the pretreatment interval (Yoshino and Kimura et al., 2006). Levels of oxidative stress marker 3-nitrotyrosine in CSF declined for most patients. 

In 2006, the company began a Phase 3 study in 206 ALS patients at 29 sites in Japan. Participants had to be able to feed themselves and show normal daily function, i.e., have no advanced disease yet. After a 12-week observation period, participants received six cycles of 60 mg edaravone or placebo over 24 weeks, followed by a 24-week open-label extension. The trial was negative on its primary endpoint, i.e., showed no significant group difference on the ALSFRS-R score, after 24 weeks of participants receiving edaravone or placebo (Abe et al., 2014). Subsequently, a post hoc analysis suggested a slowing of decline for a subset of patients with shorter disease duration and milder symptoms (Takahashi et al., 2017).

From 2006-2008, the company ran a similarly designed Phase 3 study, enrolling 25 patients with more advanced symptoms who needed help with eating and moving around. In this trial, edaravone did not affect the decline in ALSFRS-R (Writing Group, 2017).

In 2011-2014, a third Phase 3 trial ran, using strict inclusion criteria to select the subset of patients most likely to benefit, based on the prior post hoc analysis. This trial enrolled 137 patients in 29 sites in Japan. It used the same 24-week treatment schedule, with a 24-week open-label extension. The study reported less worsening on the ALSFRS-R in the edaravone group, who lost 5.01 points compared to a 7.50-point loss for placebo. There was no difference in adverse events or serious adverse events between edaravone and placebo, and no deaths. There were no positive effects on secondary findings of lung capacity, movement, survival, or other measures (Jan 2016 conference news; Writing Group et al., 2017). In a post hoc analysis of 123 patients who completed the open-label extension, slowing on the ALSFRS-R was observed by 24 weeks, and maintained at 48 weeks (Shefner et al., 2020). A separate post hoc analysis of individual ALSFRS items identified lower motor functions, such as walking and climbing stairs, as those preserved by edaravone (Brooks et al., 2022). On the basis of this study, edaravone received marketing approval in Japan, and then in the U.S. Later, an analysis of the study design questioned whether the risks of repeated infusions outweighed the drug’s apparent benefits (Turnbull, 2020).

In October 2019, two observational studies began to examine biomarkers of edaravone action. One is enrolling 160 ALS patients in the U.S. The investigators will categorize patients into four ALS phenotypes—lower-motor-neuron predominant, upper-motor-neuron predominant, bulbar predominant, generalized ALS—and compare markers of oxidative stress and antioxidant capacity in blood, urine, and CSF in those taking edaravone or not as part of their clinical care. The study will run through September 2023.

Mitsubishi Tanabe Pharma is sponsoring the second observational study. REFINE-ALS follows 300 patients prescribed edaravone, who will give blood and urine samples to be tested for markers of oxidative stress, inflammation, and neurodegeneration. This study will run through March 2023 at 39 locations in the U.S. (Berry et al., 2021).

In a meta-analysis of published post-marketing clinical data, edaravone appeared effective in Asian countries, where its reported benefits on ALSFRS-R scores and lung capacity were similar to those seen in the clinical trials. The drug appeared to have little clinical benefit in European countries. There was insufficient data in the U.S. to draw conclusions (Ortiz et al., 2020). In another study of U.S. military veterans with ALS, edaravone use was associated with a higher risk of hospitalization (Vu et al., 2020). A descriptive study of edaravone use in Argentina found adverse effects were rare, but access was limited, with fewer than half of patients covered through health insurance, and longer times to begin treatment compared to riluzole (Quarracino et al., 2020). Two studies in Japan reported improved long-term survival with edaravone (Okada et al., 2018; Houzen et al., 2021). In the latter study, median survival was 49 months in patients treated with edaravone for an average of two years, compared to 25 months for a clinically similar group who did not take edaravone. This small study enrolled 45 patients. At the April 2022 American Academy of Neurology conference, Mitsubishi Tanabe Pharma reported that, based on an analysis of U.S. insurance claim records, ALS patients who took edaravone for at least a year survived six months longer than patients who did not take the drug (press release).

A post hoc analyses of prior clinical trials, and an observational study, linked baseline urate levels and their rate of change to disease progression and treatment response (Takahashi et al., 2022Han et al., 2022).

In Italy, a small study attempting to monitor edaravone efficacy with MRI reported more cortical thinning and white-matter-tract fractional anisotrophy in the edaravone compared to control group, implying faster progression (Distaso et al., 2021). 

In January 2022, a post-marketing study of real-life use reported that edaravone had failed to slow disease progression compared to the standard treatment of riluzole (Witzel et al., 2022, editorial by Glass and Fournier, comment by Richard Bedlack). Conducted by German researchers between 2017 and 2020, the study compared outcomes for 141 ALS patients who received edaravone plus riluzole with those of 130 comparable controls who got riluzole only. After a median treatment time of 13.9 months, the groups showed no difference in decline on the ALSFRS-R, survival probability, time to ventilation, or rate of progression. This negative outcome held even for a subgroup of more mildly affected patients who met all inclusion/exclusion criteria of the positive Japanese Phase 3 trial. Adverse effects consisted mainly of infusion site infections and allergic reactions and were noted in 16 percent of participants. A small study of 30 patients in India also found no effect on ALSFRS after six months of treatment (Tomar et al., 2022).

Mitsubishi Tanabe Pharma has developed an oral edaravone formulation under the name MT-1186. In 2019-2020, MT-1186 completed safety and bioequivalence studies in Japan (Shimizu et al., 2021; Shimizu et al., 2021, Shimiziu et al., 2022). In November 2019, the company began an open-label safety study in 150 ALS patients in North America, Europe, and Japan. Participants, who must be living and functioning independently, will take the drug in the same four-week on-off cycles as the infused drug, for up to 48 weeks. The primary outcome is adverse events, with secondary outcomes to be changes in ALSFRS-R and time to death. The trial ultimately enrolled 185 patients, was completed in October 2021, and was reported to have shown good tolerability (Genge et al., 2022). The company added a 96-week extension that will run through September 2023.

In November 2020, the company started a 48-week Phase 3 efficacy study of oral edaravone at 84 locations worldwide. It aims to enroll 380 participants with a similar mild symptomatic profile as the IV Phase 3. The trial has two arms, where participants take drug daily or in on/off cycles. There is no placebo group. The primary outcome remains the ALSFRS-R. The trial is scheduled to end in July 2023; a two-year extension is planned to run until June 2024. In January 2022, two similar, 300-patient efficacy studies of the oral formulation started up. One is enrolling at 20 sites in the United States, Canada, the United Kingdom, and Japan; the other is enrolling at 38 sites across Europe; both will run until June 2024.  

The Dutch company Treeway is also developing an oral formulation of edaravone for ALS, called TW001, for ALS and Alzheimer's disease.

Edaravone is also being trialed in ischemic stroke, intracerebral hemorrhage, alcohol-induced brain injury, and optic neuritis. It appears to improve outcomes after ischemic stroke in trials in certain countries in Asia (Chen et al., 2021; Xu et al., 2021). A nanoparticle formulation reduced inflammation and improved neurological function after surgery for cerebral hemorrhage in Chinese patients (Dang et al., 2021).

For details on edaravone trials, see clinicaltrials.gov

Last Updated: 15 Dec 2022

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

PBFT02

Tools

Back to the Top

Overview

Name: PBFT02
Therapy Type: DNA/RNA-based
Target Type: Other (timeline)
Condition(s): Frontotemporal Dementia
U.S. FDA Status: Frontotemporal Dementia (Phase 1/2)
Company: Passage Bio

Background

PBFT02 is a gene-replacement therapy that uses adeno-associated virus serotype 1 (AAV1) to deliver a functional copy of the progranulin gene GRN to the brain. Progranulin mutations are a frequent cause of familial frontotemporal dementia. Mutations result in 30 to 50 percent reductions in cerebrospinal fluid progranulin levels compared to the normal range. Progranulin insufficiency interferes with proper lysosomal function. PBFT02 is intended to overcome progranulin deficiency in mutation carriers.

PBFT02 is delivered as a one-time injection into the cerebrospinal fluid in the cisterna magna at the base of the brain.

In preclinical work, intraventricular injection of AAV-GRN into progranulin-deficient mice led to elevated progranulin concentration in the brain and CSF, normalization of lysosome function, and reduction in microgliosis. In nonhuman primates, a single injection into the cisterna magna raised CSF progranulin to 40 times normal human levels, with no apparent toxic effects (Hinderer et al., 2020).

Findings

In January and March 2021, PBFT02 received Orphan Drug Designation and Fast Track status, respectively, from the U.S. FDA (company press release).

In September 2021, Passage Bio began UpliFT-D, a Phase 1/2 trial to assess safety, tolerability, and efficacy of PBFT02 in 15 people with symptomatic FTD and progranulin mutations. Participants must be between ages 35 and 75 and not live in a nursing home. The open-label study will test doses of 3.3 x 1010 and 1.1 x 1011 gene copies per gram brain weight in two sequentially enrolled cohorts, with an optional third cohort at a higher dose of 2.2 x 1011 copies per gram. Primary outcomes are adverse events, safety measures of peripheral nerve function, and antibodies to the vector and transgene in serum and CSF up to five years after administration. The 23 secondary outcomes span neurocognitive and other FTD assessments, plasma and CSF progranulin levels, biomarkers of disease progression, MRI, activities of daily living, and survival from baseline to two years. The study is running at centers in North America, Brazil, and Europe, with primary completion slated for August 2024.

In On December 20, 2023, the company announced six-month safety results from the first dose cohort (press release; May 2024 company slides). The first patient, who received low-dose steroid immunosuppressive treatment, experienced two serious adverse events of liver toxicity and blood clots in the brain. Both were asymptomatic. The patient left the study after 10 weeks. The next two patients were given higher steroid doses, and reported only mild to moderate adverse events. The investigators reported no evidence of immune response, liver toxicity, or imaging abnormalities in these two patients, who continue to be monitored. None of the three showed changes in peripheral nerve function that would indicate dorsal root ganglion toxicity. In all patients, CSF progranulin concentrations increased 3.6- to 6.6-fold over baseline after 30 days, and exceeded the levels in healthy adults. Plasma progranulin remained low.

For details on PBFT02 trials, see clinicaltrials.gov.

Last Updated: 03 Jul 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