Therapeutics

Tideglusib

Tools

Back to the Top

Overview

Name: Tideglusib
Synonyms: NP031112, Nypta® , Zentylor™ , Glycogen synthase kinase 3 inhibitor, NP12
Chemical Name: 4-Benzyl-2-(naphthalen-1-yl)-1,2,4-thiadiazolidine-3,5-dione
Therapy Type: Small Molecule (timeline)
Target Type: Tau (timeline)
Condition(s): Alzheimer's Disease, Progressive Supranuclear Palsy
U.S. FDA Status: Alzheimer's Disease (Discontinued), Progressive Supranuclear Palsy (Discontinued)
Company: Zeltia Group

Background

Tideglusib is an orally available, small-molecule drug of the thiadiazolidinone class. It acts as an inhibitor of glycogen synthase kinase 3 (GSK3-β), a widely studied tau kinase. The rationale for its use is that inhibiting GSK3-β will counteract tau hyperphosphorylation, which is thought be a step in the cellular pathogenesis that leads to Alzheimer's disease and other neurodegenerative diseases marked by neurofibrillary tau pathology. Tideglusib was being developed for the treatment of Alzheimer's and the tauopathy progressive supranuclear palsy (PSP) as part of a drug-discovery program based on marine-derived GSK3-β inhibitors. In preclinical studies, tideglusib was reported to reduce a range of disease outcomes, including tau phosphorylation, amyloid deposition, neuron loss, and gliosis in mouse entorhinal cortex and hippocampus, and to reverse a spatial memory deficit in transgenic mice. Neuroprotective, anti-inflammatory, and neurogenesis-inducing effects of tideglusib have been reported in animal models, as well (e.g. Sereno et al., 2009Morales-Garcia et al., 2012, Wang et al., 2016).

Findings

A Phase 2a, escalating-dose trial compared four escalating doses ranging from 400–1,000 mg/day to placebo in 30 patients with mild to moderate AD. Conducted at three sites in Germany, this five-month trial evaluated safety and effects on cognition and mood. It reported good tolerability except for a transient increase in serum transaminase levels. It also reported trends for a cognitive benefit on the MMSE screen and ADAS-Cog test battery (see del Ser et al., 2013).

A subsequent six-month, Phase 2b trial in 306 patients with mild to moderate AD ran in 55 centers in Spain, Germany, and other European countries. It compared 1,000 mg taken once daily, 1,000 mg taken every other day, and 500 mg once daily against placebo, and measured cognition with the ADAS-Cog as primary outcome. Called ARGO, the trial started in April 2011 and ended in July 2012. This trial was reported to have missed its primary endpoint and some secondary endpoints (see El Economista articlePharmaTimes article); subsequently, results were formally published (Lovestone et al., 2015).

In 2009, tideglusib received FDA and EMA orphan drug status, and in 2010 received FDA fast-track status for progressive supranuclear palsy, a tauopathy considered suitable to test treatments targeting this protein. In 2009, Noscira started a one-year study in 146 patients with PSP. Called TAUROS, this multicenter European trial compared 600 and 800 mg of tideglusib taken once a day to placebo, measuring change in the Progressive Supranuclear Palsy Rating Scale of Golbe as primary outcome. Tideglusib reportedly showed a signal toward reduced brain atrophy in a subgroup analysis (see company press release, Hoglinger et al., 2014); however, this trial, too, was negative on the primary outcome (Tolosa et al., 2014)

In 2016, clinial trials started evaluating tideglusib for the treatment of congenital As of 2019, tideglusib was in clinical trials for myotonic dystrophy. For all cinical trials of this drug, see clinicaltrials.gov.

Noscira was set up in 2000 as a subsidiary of the Spanish Zeltia Group, under the name Neuropharma. It was renamed Noscira in 2008, and its impending liquidation was announced in December 2012 (see Reuters story).

Last Updated: 03 Jun 2019

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

ORM-12741

Tools

Back to the Top

Overview

Name: ORM-12741
Therapy Type: Small Molecule (timeline)
Target Type: Other Neurotransmitters (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: Orion Pharma

Background

ORM-12741 is an orally available α 2c adrenergic receptor antagonist being developed for Alzheimer's disease by the Finnish company Orion. The role of the α 2c adrenergic receptor in Alzheimer's disease is poorly understood. Its role in the cognitive deficits of schizophrenia is better-studied (Kalkman and Loetscher, 2003). ORM-12741 was originally found as part of a schizophrenia drug discovery program, but after some early clinical studies in Europe is not currently being developed for this indication.  The α 2c adrenergic receptor modulates numerous brain systems, including brain activity during stress. No peer-reviewed studies about ORM-1274 have been published, but according to Orion, the compound improved memory function in rodent studies (Orion media release).

Findings

From 2008 to 2010, Orion Pharma had conducted seven Phase 1 trials for ORM-12741 in a total of more than 200 healthy volunteers in Finland, France, and the Netherlands. These studies evaluated absorption, distribution, metabolism, and excretion (ADME) of 14C-labelled ORM-12741, indicating that a PET tracer is available to accompany clinical development of this drug and evaluate blood-brain barrier penetration and receptor occupancy, for example. Other Phase 1 trials examined safety, pharmacokinetics, food effects, and other parameters of single and multiple ascending doses.

From April 2011 to October 2012, a three-month Phase 2a trial in Finland and Spain compared flexible doses of 30 to 60 or 100 to 200 mg of ORM-12741 to placebo in 100 patients with moderate Alzheimer's disease who were already taking cholinesterase inhibitors or memantine. The trial assessed safety via adverse events, vital signs, laboratory values and ECG, and well as efficacy via computerized cognitive tests of various memory domains. Data were presented at the 65th Annual Meeting of the American Academy of Neurology held March 2013 in San Diego, and again at the World Congress of Neurology in September 2013 in Vienna. ORM-12741 was reported to have been generally well-tolerated and to have shown small but statistically significant benefits on working memory, episodic memory, and caregiver distress (for details, see AAN postercompany press release).

In June 2015, Orion started a Phase 2 study, called Nebula, to evaluate the efficacy of ORM-12741 for calming agitation and aggression in Alzheimer's disease. The study compares a 12-week course of either a low dose or high dose twice daily to placebo. It enrolls 300 people with probable Alzheimer's disease of moderate severity, and clinically significant agitation and aggression for change on the NPI clinician rating scale plus a range of secondary outcomes. This trial takes place in Germany, the Czech Republic, Poland, Finland, Bulgaria, and Croatia, and is set to run until summer 2017.

Previous Phase 2 clinical development of this drug for Raynaud's phenomenon, a condition marked by cold-induced vasospasms, was terminated in January 2013 following a recommendation by the trial's Data and Safety Monitoring Committee (see NCT01315899). For all clinical trials of this drug listed in the United States see clinicaltrial.gov and EU Clinical Trials Register.

Clinical Trial Timeline

  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034
Orion Pharma NCT01324518
N=100

Last Updated: 18 Nov 2016

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Minocycline

Tools

Back to the Top

Overview

Name: Minocycline
Synonyms: Solodyn, Arestin, Minocin, Dynacin
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)

Background

This second-generation tetracycline antibiotic has been in use for more than 30 years. It also has anti-inflammatory activity, crosses the blood-brain barrier, and inhibits microglia activation. Based on these properties, and the central role of microglia in Alzheimer’s, this drug was considered a repurposing candidate for evaluation in AD (Corbett et al., 2015; Appleby and Cummings 2013).

Preclinically, minocycline prevented Aβ fibrillization and Aβ-induced microglial activation in vitro (Familian et al. 2006). In mouse and rat models of AD, minocycline inhibition of microglial activation led to reduced amyloid and tau deposition, protection of hippocampal neurogenesis, and improved cognition (Seabrook et al. 2006; Choi et al., 2007; Nobel et al., 2009; Biscaro et al., 2012).

Minocycline displayed neuroprotective activity in preclinical models of multiple other neurologic conditions, but subsequently failed to show efficacy in clinical trials for Huntington’s and Parkinson's diseases, multiple systems atrophy, schizophrenia, multiple sclerosis, and traumatic brain injury.  In people with amyotrophic lateral sclerosis, minocycline accelerated deterioration (Gordon et al., 2007).

Findings

In May, 2014, a Phase 2, multicenter trial began enrolling people with mild AD from National Health Service memory clinics in Scotland and England. Participation required a clinical diagnosis and a standardized MMSE score above 24. The 544 participants took 200 or 400 mg minocycline per day, or placebo, for two years. The primary outcomes in this pragmatic trial were change from baseline in MMSE and Bristol Activities of Daily living, assessed every six months.

Full trial results are published (Nov 2019 news on Howard et al., 2019). After 24 months, minocycline on neither dose slowed cognitive or functional decline relative to placebo. The high dose was poorly tolerated: around 60 percent of the low-dose and placebo groups completed the trial, fewer than 30 percent of the high-dose group did. The most common reasons for withdrawal were gastrointestinal side and skin problems, and dizziness.

This trial was listed in the EU Clinical Trials Register. For more trials on minocycline, see clinicaltrials.gov.

Last Updated: 06 Dec 2019

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

SAR110894D

Tools

Back to the Top

Overview

Name: SAR110894D
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: Sanofi

Background

Last Updated: 10 Aug 2013

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Rilapladib

Tools

Back to the Top

Overview

Name: Rilapladib
Synonyms: SB-659032
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: GlaxoSmithKline (GSK)

Background

Rilapladib is an inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2). Also known as plasma platelet-activating factor acetyl hydrolase, this enzyme is released into the blood by monocytes/macrophages. Lp-PLA2 can have pro-inflammatory and oxidative properties; it is associated with LDL cholesterol and linked to the formation of atherosclerotic plaques. Rilapladib was originally developed for atherosclerosis, but GSK discontinued this indication in Phase 2.  

Lp-PLA2 drew interest as a target in Alzheimer's disease when research into the brain's lipid metabolism identified it as a potential factor in cerebrovascular oxidative stress and inflammation. Some studies implicated Lp-PLA2 produced locally in vascular plaques with endothelial dysfunction (Lavi et al., 2007Adibhatla and Hatcher, 2008). Another Lp-PLA2 ihibitor, darapladib, lowered the total amount of brain Aβ in diabetic and hypercholesterolemic pigs (Acharya et al., 2013).

The epidemiological evidence is mixed. Some studies have linked Lp-PLA2 to increased risk for ischemic stroke and developing dementia (Oei et al., 2005van Oijen et al., 2006Fitzpatrick et al., 2014).  However, the Framingham Heart Study was unable to associate Lp-PLA with risk for dementia, or with microbleeds as indicators of cerebral amyloid angiopathy (van Himbergen et al., 2012Romero et al., 2012).  Genetic association studies of Lp-PLA2 have been largely negative. 

A case-control study of cognitively normal and cognitively impaired people reported no association between plasma Lp-PLA2 levels and cognition (Davidson et al., 2012). In similarly designed studies, higher activity of Lp-PLA2 was identified in people with vascular dementia, but not Alzhiemer’s (Zuliani et al., 2023; Savas et al., 2016). Other studies found higher activity associated with AD, possibly by way of vascular damage (Bacchetti et al., 2015; Doody et al., 2015).

Findings

In October 2011, GSK started enrolling 124 people with mild to moderate Alzheimer's disease into a Phase 2 study comparing 250 mg of rilapladib taken once daily to placebo. This study primarily evaluated rilapladib's effect on CSF Aβ and tau biomarkers, as well as on a composite score of working memory and executive function. The study ended in February 2013, and results have been published (Maher-Edwards et al., 2015). Rilapladib produced an improvement in the executive function/working memory composite at week 24, but no change in CSF Aβ42 or other endpoints.

In 2014, the company listed a small Phase 1 study enrolling 30 healthy volunteers to look at pharmacokinetics and elimination of rilapladib, and separately at the effect of the antifungal drug itraconazole on rilapladib. This study was withdrawn before enrollment started.

In 2022, Shanghai-based SciNeuro announced it had bought the rights to rilapladib and other Lp-PLA2 inhibitors (press release). In March 2023, SciNeuro began Phase 1 with SNP318, a next-generation inhibitor claimed to have better CNS penetration than rilapladib.

See all rilapladib trials on clinicaltrials.gov

Last Updated: 21 Sep 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Nasal Insulin

Tools

Back to the Top

Overview

Name: Nasal Insulin
Synonyms: Detemir, Levemir, Humulin, Novolin, glulisine
Therapy Type: Small Molecule (timeline), Other
Target Type: Amyloid-Related (timeline), Other (timeline)
Condition(s): Alzheimer's Disease, Mild Cognitive Impairment, Parkinson's Disease, Multiple System Atrophy
U.S. FDA Status: Alzheimer's Disease (Phase 2/3), Mild Cognitive Impairment (Phase 2), Parkinson's Disease (Phase 2), Multiple System Atrophy (Phase 2)
Approved for: Diabetes

Background

Insulin is the peptide hormone that is standard treatment for the management of Type 1 diabetes. In a new therapeutic approach, various forms of commercially available insulin are being atomized into a spray and inhaled through the nose. The rationale behind this exploration of insulin for the treatment of cognitive impairment and dementia is twofold. First, brain areas affected in Alzheimer's disease have been shown to express insulin receptors, and insulin levels as well as insulin receptor signaling are thought to be reduced in Alzheimer's (e.g., Chiu et al., 2008Steen et al., 2005). Second, if effective, delivery of this hormone directly into the brain along olfactory perivascular channels would sidestep the peripheral bloodstream. This would avoid the unwanted effect of increasing systemic insulin levels, which could lead to hypoglycemia or insulin resistance (e.g., Born et al., 2002).

The physiological role of insulin in the brain is incompletely understood, but it has been implicated in synaptic remodeling and glucose utilization. Insulin has also been proposed to protect against Aβ toxicity, and insulin deficits have been linked to tau pathology, neuroinflammation, and other aspects of Alzheimer's pathophysiology (De Felice et al., 2009; El Khoury et al., 2014Zhao et al., 2009). Insulin resistance, Type 2 diabetes, obesity, and metabolic syndrome are risk factors for Alzheimer's disease (see AlzRisk Diabetes, AlzRisk Obesity).

Intranasal insulin began attracting attention in Alzheimer's research when small human studies reported improved cognition without a change in blood glucose or insulin levels in healthy volunteers (e.g., May 2002 news; Benedict et al., 2004Benedict et al., 2007).

Findings

A single-center pilot study at the Veterans Affairs Hospital/University of Washington in Seattle reported improved verbal memory retention and attention after a three-week test of 20 international units (IU) daily of intranasal insulin compared with placebo in 24 people with amnestic memory impairment (aMCI) or mild Alzheimer's disease. The insulin was delivered with an electronic atomizer (Reger et al., 2008).

A separate small trial comparing different doses in 33 people with aMCI or early AD reported differential effects by ApoE genotype (Reger et al., 2008).

Intranasal insulin appeared safe in a subsequent four-month course of 20 or 40 IUs of intranasal insulin or placebo given to a larger group of 104 people with aMCI or mild to moderate AD. Called SNIFF-120, this single-center study reported some preservation of cognition in younger participants and some preservation of function. The study reported no change in the CSF biomarkers between the groups, but some signals on subgroup analysis (Craft et al., 2012; Jul 2010 news). Different responses by gender or ApoE genotype were also reported (Claxton et al., 2013).

In September 2013, the Alzheimer's Disease Cooperative Study began enrolling for Study of Nasal Insulin in the Fight Against Forgetfulness (SNIFF), a federally funded study that delivered 20 IU of insulin or placebo after breakfast and dinner to 289 people with either aMCI or early AD. The first 49 participants used the same insulin delivery device as had been used in the previous trial; the remaining 240 used a different one. After one year of treatment, there was no cognitive or functional difference between insulin and placebo groups in the cohort of 240 who used the second delivery device. The smaller group of 48 who used the original device did show slowing of worsening in the ADAS-COG-12 subscale and activities of daily living at one year (Craft et al., 2020). A secondary analysis of MRI data from this smaller group found reduced accumulation of white-matter abnormalities in treated compared to placebo patients after one year (Kellar et al., 2021). Subsequent measurement of CSF markers of inflammation, immune function, and vascular integrity in these participants suggested that the observed clinical benefit correlated with changes consistent with anti-inflammatory properties of insulin (Kellar et al., 2022).

At AD/PD in March 2021, additional trial results were presented. Participants using the original device continued to do better than those on placebo during a six-month open-label period. There was no effect of insulin in the larger cohort using the second device. In a subsequent Phase 2 study to determine how much insulin entered the brain after nasal delivery, 19 people received 40 IU insulin or a placebo using the first device, followed by lumbar puncture. CSF insulin concentrations approximately doubled after 30 minutes. The manufacturer of the second device refused to allow its testing.

In July 2020, a Phase 2 trial of similar design began to compare three different intranasal delivery devices and two doses of insulin, 20 and 40 IU, in 30 people with no or mild cognitive impairment. It will run until October 2023.

In October 2021, a Phase 2 trial started to test nasal insulin alone or in combination with the insulin sensitizer empaglifozin. Taken in pill form, empagliflozin was recently shown to improve the response to nasally delivered insulin in the hypothalamus in people with prediabetes (Kullmann et al., 2022). The single site study at Wake Forest University will enroll 60 participants with elevated brain amyloid and normal cognition, MCI, or early dementia. The four-arm protocol will compare nasal insulin four times daily, one 10 mg empagliflozin pill daily, or both, to placebo. Safety is the primary outcome; secondaries include cognitive measures, CSF Aβ and tau, and brain blood flow. The study will run until 2028.  

This trial will use the Aptar Pharma CPS Intranasal Delivery Device, which is different from devices in previous SNIFF studies. A trial, planned to begin in May 2022, will assess adherence and CSF insulin concentrations in 60 people with AD or MCI who use the device for three weeks. The placebo-controlled study will also measure cognitive, functional, and biomarker outcomes. The estimated end date is May 2026.

The trials of intranasal insulin vary in that some evaluate fast-acting forms of insulin as used in diabetes therapy, while others evaluate longer-acting insulin analogs. For example, two trials conducted at the University of Washington, Seattle, between 2011 and 2013 evaluated insulin detemir, a long-acting insulin analog that differs from human insulin by one amino acid. A three-week study in 60 patients with MCI or AD compared 10 and 20 IUs of detemir twice daily to placebo. The higher dose improved memory in ApoE4 carriers, worsened it in noncarriers, and did not change daily function or executive function (Claxton et al., 2015). A subsequent four-month study compared 20 IUs twice daily of insulin detemir to 20 IUs of regular insulin twice daily in 36 patients. Regular insulin, but not detemir, improved memory compared with placebo, and was associated with preserved brain volume in several regions on MRI (Craft et al., 2017). These trials did not measure exposure or target engagement.

In a small study examining glulisine, a rapid acting form of insulin, nasal delivery of 20 UI twice daily for six months failed to improve cognition, function, or mood compared to placebo in 35 people with MCI or mild probable AD (Rosenbloom et al., 2021). In pilot trials, the same investigators reported no acute improvement in cognition after a single dose of glulisine compared to placebo in nine people with AD, or 12 with Down's syndrome (Rosenbloom et al., 2014Rosenbloom et al., 2020). From 2015 to 2019, a pilot study of rapid-acting insulin aspart took place at Wake Forest. Twenty-four participants with AD got 20 UI twice daily for 12 weeks, against a primary outcome of change in the ADAS-Cog MCI scale. The trial finished in 2019.

Nasal insulin is also being evaluated for neurologic conditions other than Alzheimer's. In February 2014, a trial started up at the University of Massachusetts. It evaluated the effect of 40 IUs of regular insulin on a visuospatial memory test, the Unified Parkinson's Disease Rating Scale, and other measures compared to placebo in 15 patients with Parkinson's disease. Insulin improved verbal fluency and clinical measures of PD severity compared to placebo. One patient with MSA remained stable on insulin treatment (Novak et al., 2019).

Furthermore, various trials have evaluated nasal insulin for effects on cognitive symptoms in psychiatric conditions such as schizophrenia, bipolar, and major depressive disorder. 

For trials on nasal insulin in Alzheimer's, see clinicaltrials.gov

Last Updated: 04 Mar 2022

Further Reading

No Available Further Reading

Therapeutics

MSDC-0160

Tools

Back to the Top

Overview

Name: MSDC-0160
Synonyms: Mitoglitazone
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: Metabolic Solutions Development Company

Background

Last Updated: 10 Aug 2013

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Circadin

Tools

Back to the Top

Overview

Name: Circadin
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2)
Company: Neurim Pharmaceuticals Ltd.
Approved for: Insomnia

Background

Circadin is an extended-release formulation of melatonin, a hormone that is released by the pineal gland into the CSF at night to regulate circadian rhythms and induce sleepiness. Circadin is being marketed for insomnia in the European Union, Israel, Australia, and countries in Asia and elsewhere. It has been approved in these countries for this indication since 2007, and is in Phase 3 evaluation for sleep disorders in the United States. 

Poor sleep has been linked to increased risk of cognitive dysfunction and Alzheimer's (Miyata et al., 2013Lim et al., 2013Ju et al., 2014). Melatonin release decreases with age and becomes further dysregulated in AD (Mishima et al., 1999). Alzheimer's disease is frequently accompanied by sleep disturbances, but some sleep medications are counter-indicated because they may increase risk of dementia (Billoti de Gage et al., 2012Wu et al., 2009). 

Findings

One trial of Circadin 2 mg, as add-on to standard therapy, has been conducted in Alzheimer's disease patients. Starting in 2009 at five sites in the United States and the United Kingdom, 73 male and female outpatients with mild to moderate AD, with and without diagnosed insomnia, were randomized to a nightly pill of Circadin or placebo for six months, then switched to two weeks of washout with placebo. The primary outcome was cognitive function measured by ADAS-cog; secondary outcomes were global function as measured by IADL and sleep efficiency by way of the Pittsburgh Sleep Quality Index (PSQI). Published results report no treatment benefit on ADAS-cog, but do report a treatment benefit on IADL and on MMSE. PSQI showed no treatment effect in the whole group, but did show a treatment benefit in the co-morbid insomnia subgroup. This small subgroup reportedly showed larger, significant treatment effects on all measures (Wade et al., 2014). 

For details on this trial, see clinicaltrials.gov, for all clinical trials with Circadin, see clinicaltrials.gov

Last Updated: 09 Oct 2015

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

EVP-0962

Tools

Back to the Top

Overview

Name: EVP-0962
Synonyms: EVP 0015962
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease, Mild Cognitive Impairment
U.S. FDA Status: Alzheimer's Disease (Discontinued), Mild Cognitive Impairment (Discontinued)
Company: FORUM Pharmaceuticals Inc.

Background

EVP-0962 is a γ-secretase modulator. The rationale behind this approach is that γ-secretase modulators affect the enzyme complex differently than inhibitors: They shift its processive cleavage mechanism away from generating Aβ42 and other long forms of this peptide and toward generation of Aβ38 and other short forms of Aβ. Gamma-secretase modulators are designed to selectively reduce the amount of Aβ42 while letting cleavage of the Notch substrate proceed unimpeded. Aβ42 is the most aggregation-prone form of the peptide and lies upstream of the amyloid cascade. Inhibition of Notch cleavage is thought to have been largely responsible for the toxic side effects on the skin, immune, and gastrointestinal systems that occurred with γ-secretase inhibitors. In addition, γ-secretase inhibition does not affect the total amount of Aβ in the brain.

EVP-0962 is an NSAID derivative without activity on the classic NSAID targets Cox 1 or 2. In preclinical studies, EVP-0962 was reported to decrease Aβ42, increase Aβ38, but leave the total Aβ load or Notch processing unchanged in four different cell types. In the brains of wild-type and Tg2576 mouse models of Alzheimer’s amyloidosis, it reduced Aβ42, neuroinflammation measured as astrocyte and microglial activation, and plaque load in the hippocampus. EVP-0962 reversed memory deficits in contextual fear-conditioning tests. All effects were reported to be dose-dependent (see Apr 2011 conference story; Rogers et al., 2012).

Findings

In 2011, FORUM Pharmaceuticals Inc. (formerly called EnVivo Pharmaceuticals), ran an ascending single- and multiple-dose Phase 1 study to assess safety, tolerability, pharmacology, and food effect of EVP-0962 in healthy volunteers (see company press release).

In November 2012, FORUM Pharmaceuticals started a single-center Phase 2 study in 52 participants. This is a two-part study. The first part evaluates the safety, tolerability, pharmacokinetics, and pharmacodynamics of escalating EVP-0962 doses of 10, 50, 100, and 200 mg taken once daily for two weeks in healthy participants. The data from this part of this trial will serve to select one or several doses for part two. It will evaluate the same parameters following once-daily administration for two weeks in patients with mild cognitive impairment due to AD as defined in the NIA-AA proposed criteria. Adverse events are a primary outcome of this study, as are the rate of synthesis and concentration of CSF Aβ in response to EVP-0962 monitored for 36 hours after dosing.

Clinical Trial Timeline

  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034
FORUM Pharmaceuticals Inc. NCT01661673
N=52

Last Updated: 18 Dec 2020

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Nelonicline

Tools

Back to the Top

Overview

Name: Nelonicline
Synonyms: ABT-126
Therapy Type: Small Molecule (timeline)
Target Type: Cholinergic System (timeline)
Condition(s): Alzheimer's Disease, Schizophrenia
U.S. FDA Status: Alzheimer's Disease (Discontinued), Schizophrenia (Inactive)
Company: AbbVie

Background

ABT-126 is an α-7 nicotinic acetylcholine receptor (α7-nAChR) allosteric modulator that was being developed to treat cognitive deficits in schizophrenia and Alzheimer's disease. Cholinergic function declines in Alzheimer's, and currently approved acetylcholinesterase-inhibitor therapies modestly improve cognitive deficits in patients with AD by way of boosting cholinergic transmission. The rationale of α7-nAChR agonists was that they will enhance cognition without causing side effects associated with overactivation of other nAChRs or muscarinic AChRs. ABT-126 was initially developed by Abbott Laboratories. In January 2013, Abbott spun out its pharmaceuticals business into a new company called AbbVie.

Findings

ABT-126 was in Phase 2 development for Alzheimer's disease in the United States, Canada, South Africa, and Eastern European countries, and in Phase 3 development for cognitive symptoms in schizophrenia in the United States and Europe. 

For Alzheimer's disease, Phase 1 research for safety and pharmacology parameters started in healthy volunteers in 2009, and in patients with mild to moderate Alzheimer's in 2011. Also in 2009, a 12-week Phase 2 study in 274 people with mild to moderate AD compared 5 mg and 25 mg of ABT-126 to placebo and to donepezil, but without co-administration of ABT-126 and donepezil. This trial ended in 2010, and in 2013 preliminary data was reported to have shown good tolerability for ABT-126, with side effects similar to donepezil. A cognitive benefit for the higher dose similar to that seen with donepezil was reported as well. The cognitive signal correlated with blood measures of exposure to ABT-126 (see Aug 2013 conference news). Full data were subsequently published open-access (Gault et al., 2015). 

In 2012, AbbVie started two 24-week Phase 2b trials in 400 patients each, comparing doses ranging from 25 to 75 mg. One trial compared ABT-126 added on to treatment with an acetylcholinesterase inhibitor to placebo, the other compared ABT-126 monotherapy to placebo. Preliminary results were published, suggesting that efficacy at these higher doses was insufficient to continue further development (Gault et al., 2014, and Othman et al., 2014). Each trial offered a 28-week, open-label extension to people who had completed the blinded portion of the study. These studies were due to read out in 2014, but have been terminated. In January 2016, results of the add-on study were formally published, showing insufficient efficacy to continue development; similarly, in October, a peer-reviewed paper on the results of the monotherapy trial showed no significant improvement with any of the ABT-126 doses tested (Florian et al., 2016, Gault et al., 2016).

In schizophrenia, a 2009 Phase 1 trial evaluated safety and pharmacokinetics in 16 adult patients who were receiving treatment with an atypical antipsychotic.  In 2010, a Phase 2 study compared two undisclosed doses to placebo in 207 adults with the disease. Two additional Phase 2 trials were added, one to compare two doses to placebo in 150 patients for 12 weeks at sites in the United States; the other as an international trial to compare 25, 50, and 75 mg doses to placebo in 430 patients for 24 weeks, with a one-year open-label extension. In November 2016, a peer-reviewed paper reported no statistically significant treatment benefit for ABT-126 in the larger trial (Haig et al., 2016).

For a listing of ABT-126 trials, see clinicaltrials.gov.

Clinical Trial Timeline

  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034
AbbVie NCT00948909
N=274
AbbVie NCT01527916
N=400
AbbVie NCT01549834
N=420
AbbVie NCT01676935
N=350
AbbVie NCT01690195
N=350

Last Updated: 11 Nov 2016

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