Overview
Name: Rosiglitazone
Synonyms: Rosiglitazone maleate, Avandia
Chemical Name: 5-[[4-[2-(methyl-pyridin-2-yl-amino)ethoxy]phenyl]methyl]thiazolidine-2,4-dione, AVANDIA®
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
Rosiglitazone is an antidiabetic medication that lowers blood glucose by sensitizing pancreatic target cells to insulin. Rosiglitazone belongs to the thiazolidinedione class of peroxisome-proliferator activated receptor γ agonists; PPARγ is a nuclear receptor predominantly expressed in adipose tissue, whose activation influences expression of genes involved in glucose and lipid metabolism. The FDA approved rosiglitazone for treatment of Type 2 diabetes in 1999, but side effects soon started being reported (Sep 2003 news) and their use was restricted in 2010 after having been linked to heart attacks and strokes (see New York Times article).
PPARγ has come to be a target of interest for Alzheimer's drug development because of an overlap of metabolic disease and Alzheimer's disease risk factors. Cell-based and animal studies have implicated PPARγ, in particular, as playing a role in neuroinflammatory processes in neurodegenerative conditions. For example, PPARγ activation has been shown to modulate the microglial response to amyloid deposition in such a way that it increases Aβ phagocytosis and decreases cytokine release (see Dec 2012 news story; Mandrekar-Colucci et al., 2012).
Rosiglitazone has been variously reported to improve behavioral outcomes, normalize brain glucocorticoid receptor levels, activate wnt signaling, and decrease insoluble Aβ42 levels in mutant human APP/PS transgenic mice (Pedersen et al, 2006; Escribano et al., 2009; Toledo and Inestrosa, 2010; O'Reilly and Lynch, 2011). It was also reported to stimulate generation of mitochondria in mouse brain, and to protect synapses in hippocampal slice cultures against Aβ oligomer toxicity by way of increasing the number of mitochondria in neuronal spines and dendrites (Strum et al., 2007; Xu et al., 2013). Anti-inflammatory and anti-tau hyperphosphorylation effects have also been proposed (Xu et al. 2014; Yu et al., 2014).
Findings
A dozen clinical studies have evaluated rosiglitazone in AD.
Starting in 1999, a pilot study at the Veterans Affairs/University of Washington tested a six-month course of 4 mg per day of rosiglitazone or placebo in 30 people with amnestic MCI or mild AD. It reported a treatment benefit on recall and attention, as well as stable plasma Aβ in the treatment group versus declining plasma Aβ in the placebo group, claiming a slowing of disease progression (Nov 2002 news; Watson et al., 2005).
Subsequently, GSK ran four Phase 1 trials in a total of 142 patients to assess drug interactions, an extended-release formulation, and pharmacokinetics. From May 2004 to July 2008, GSK conducted a multicenter Phase 2 study in North America and the United Kingdom. It evaluated the effect of a one-year course of 8 mg per day of rosiglitazone on global and regional functional brain activity and cognition in 80 people with mild to moderate Alzheimer's disease. This trial reported an early increase in glucose metabolism, but no evidence for a slowing of progression at one year (Tzimopoulou et al., 2010).
From January 2004 to May 2005, GSK ran a large Phase 2 trial at 67 centers in Europe and New Zealand, comparing 2 and 8 mg of rosiglitazone per day to placebo in 511 patients with mild to moderate AD for their ability to change baseline ADAS-cog or Clinician's Interview-Based Impression of Change plus caregiver input (CIBIC+) levels. The trial missed its primary endpoint, but a subgroup analysis found a treatment benefit on the higher dose in ApoE4 non-carriers, prompting a pharmacogenomic Phase 3 program (Sep 2006 news; Risner et al., 2006).
In July 2006, GSK started two 54-week Phase 3 studies of add-on therapy in people with mild to moderate Alzheimer's disease. One compared 2 and 8 mg per day of rosiglitazone plus donepezil to donepezil alone in 1,496 participants, the other compared the same doses of rosiglitazone added to any of the three cholinesterase inhibitors approved for AD in 1,485 participants. Both these multinational trials stratified participants into ApoE4 carriers and non-carriers. ADAS-cog and CDR-SB served as co-primary outcome measures. Both trials were negative. Rosiglitazone showed no effect on cognition or function with either dose, or in any ApoE genotype (Harrington et al., 2011).
Between February 2007 and September 2008, a third Phase 3 trial compared a six-month course of either 2 mg/day or 8 mg/day rosiglitazone monotherapy to donepezil treatment in 693 people with mild to moderate AD. The ADAS-cog and CIBIC+ input were co-primary endpoints. This trial, too, was negative, with no significant difference between the groups on either outcome (Gold et al., 2011). In all Phase 3 trials, peripheral edema was the most significant adverse effect.
GSK subsequently terminated two open-label extension trials, and discontinued evaluating rosiglitazone for an indication in Alzheimer's disease.
Last Updated: 11 Mar 2016
Further Reading
No Available Further Reading
Overview
Name: Rofecoxib
Synonyms: Vioxx™
Therapy Type: Small Molecule (timeline)
Target Type: Inflammation (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Merck
Approved for: Withdrawn from market
Background
The NSAID rofecoxib is a selective cyclooxygenase-2 (COX-2) inhibitor. Its anti-inflammatory and analgesic effects appear to result from the inhibition of prostaglandin synthesis. In 2004, Merck voluntarily withdrew rofecoxib amid controversy over cardiovascular side effects (Sep 2004 news story).
In Alzheimer's research, interest in NSAIDs arose when epidemiological studies started reporting lower rates of Alzheimer's disease or protection of cognition among people who had been taking these drugs for chronic treatment of inflammatory conditions (e.g., Mar 1997 news story; in't Veld et al., 1998; Nov 2001 news story; Vlad et al., 2008; Obermann et al., 2013). Experimental studies supported the argument that inflammation plays a role in Alzheimer's pathogenesis, prompting a wave of clinical trials of various NSAIDs, such as ibuprofen, naproxen celecoxib, and R-flurbiprofen.
Findings
Three clinical trials of rofecoxib in Alzheimer's disease have been conducted. In 2000 and 2001, the Alzheimer's Disease Study Group conducted a 40-center trial that compared one year of treatment with 25 mg once daily of rofexocib or 220 mg twice daily of naproxen to placebo in 351 people with mild to moderate Alzheimer's disease. The trial assessed whether these drugs would slow cognitive decline, but found that neither showed any consistent benefit over placebo (Jul 2002 conference story; Aisen et al., 2003).
In 2004, Merck scientists published results of a larger, company-sponsored trial of of the same once-daily dose of 25 mg rofecoxib or placebo, given for one year, in 692 people with mild to moderate Alzheimer's disease. In this study, too, rofecoxib showed no effect on either cognitive or clinical/functional outcomes (Reines et al., 2004).
Similarly, a trial evaluating whether rofecoxib could delay a diagnosis of Alzheimer's disease in 725 people with mild cognitive impairment was negative, as well (see Thal et al., 2005; Aisen, 2005; and Aisen et al., 2008).
Last Updated: 25 Oct 2023
Further Reading
No Available Further Reading
Overview
Name: RG3487
Synonyms: RO5313534, MEM 3454
Chemical Name: N-[(3S)-1-azabicyclo[2.2.2]oct-3-yl]-1H-indazole-3-carboxamide hydrochloride
Therapy Type: Small Molecule (timeline)
Target Type: Cholinergic System (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Inactive)
Background
RG3487 is a selective nicotinic alpha-7 receptor (α7nAChR) partial agonist. It also has properties of a serotonin 3 receptor antagonist. It has been tested in Alzheimer's disease and cognitive symptoms of schizophrenia. In vivo, RG3487 improves object recognition memory and reverses spatial learning deficits in age-impaired rats. RG3487 improved drug-induced deficits in the prepulse inhibition, or startle, model of sensorimotor gating, and reversed impairments in executive function tasks. (Wallace et al., 2011).
Findings
Roche licensed this therapeutic from Memory Pharmaceuticals Corp. In 2006, Memory Pharmaceuticals had announced positive preliminary cognitive data from a phase 1 trial of MEM 3454, and in 2007 announced positive phase 2a results from a 2-month proof-of-concept trial in 80 Alzheimer's patients (see company press release). In 2008, Roche and Memory Pharmaceuticals expanded the development program for Mem4354 and Roche acquired Memory Pharmaceuticals (see press release).
In 2009 and 2010, Roche ran a phase 2 dose-ranging study in 389 people with mild-to-moderate Alzheimer's disease compared 6 months of treatment with 1, 5, or 15 mg/day of RG3487 to placebo. This study assessed efficacy and safety of the compound added to donepezil. Results were not reported publicly.
Also in 2010, Roche conducted an open-label phase 1 study in 16 healthy adults to evaluate the effect of memantine on the pharmacokinetics, safety and tolerability of RO5313534 and vice versa.
Development of this drug has been discontinued. For details on clinical trials on this compound, see clincialtrials.gov
Last Updated: 25 Oct 2023
Further Reading
No Available Further Reading
Overview
Name: Resveratrol
Synonyms: trans-3,4',5-trihydroxystilbene
Therapy Type: Small Molecule (timeline), Supplement, Dietary (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease, Mild Cognitive Impairment
U.S. FDA Status: Alzheimer's Disease (Phase 3), Mild Cognitive Impairment (Phase 4)
Background
Resveratrol is one of many bioactive polyphenols in certain foods, such as red grapes, blueberries, peanuts, and dark chocolate. It has been reported to have neuroprotective effects in cell-based and animal models. These were attributed to resveratrol's antioxidant capacity, or to specific processes such as prevention of amyloid deposition (Karuppagounder et al., 2009) or induction of the sirtuin 1 gene to increase translation of the anti-aging enzyme sirtuin (Kim et al., 2007). Besides Alzheimer's disease, resveratrol has been studied for the treatment of cardiovascular and metabolic disease, cancers, as well as pain, inflammation, and other conditions of aging (Albani et al., 2010; Li et al., 2012).
Resveratrol is available as part of a healthy diet or in the form of dietary supplements. Epidemiological research has questioned whether the dose achievable through diet has specific health benefits in aging (see May 2014 news). Resveratrol supplementation achieves higher blood levels, but resveratrol's low brain penetrance may limit its potential to treat Alzheimer's disease. Clinical studies of resveratrol supplementation continue across a range of indications, while proprietary small-molecule derivatives with favorable pharmacokinetic properties are being developed, as well.
Findings
Between 2008 and 2010, a Phase 3 trial at the Veterans Affairs Medical Center in the Bronx, New York, evaluated a one-year course of treatment with a liquid formulation of resveratrol, glucose, and malate in 27 patients with mild to moderate Alzheimer's. The primary outcome was change on the ADAS-cog. This study is not yet published.
In 2010, a study at the Charite University Medical Center in Berlin began a Phase 4 study in 330 people with mild cognitive impairment (MCI). This trial evaluates resveratrol supplementation as one of six different interventions against ADAS-cog as the primary outcome and brain imaging and plasma biomarkers as secondary outcomes. This trial is set to run through 2014.
In 2013 and 2014, the Alzheimer's Disease Cooperative Study conducted a 26-center Phase 2 study across the United States to evaluate a one-year course of treatment with up to 2 grams of resveratrol capsule supplementation per day in 119 people with mild to moderate Alzheimer's. Primary outcomes were change in CSF Aβ and tau biomarkers, safety and tolerability, and change in brain and hippocampal volume as measured by MRI. This trial includes a pharmacokinetic substudy that will measure resveratrol levels over a 24-hour period in 15 participants. At CTAD 2014, and in a peer-reviewed paper later on, this trial was reported to have shown that about 1 percent of resveratrol reaches the CSF. Both plasma and CSF Aβ40 levels decreased more in the placebo than the treatment group, whereas brain atrophy was reportedly greater in the treatment versus placebo group (see discussion in Dec 2014 conference news; Turner et al., 2015).
In 2014, a small study of 46 cognitively healthy, overweight people reported that a six-month course of 200 mg of resveratrol per day taken as a supplement improved both word recall and two brain imaging measures (see Jun 2014 news).
In June 2015, a Phase 1 trial at Johns Hopkins University began evaluating three doses of a nutraceutical preparation containing resveratrol and other polyphenols in 48 prediabetic people with amnestic MCI.
Other small clinical studies have investigated the effect of resveratrol supplementation on cerebral blood flow in healthy adults, on the neurodegenerative disease Friedreich's ataxia, and on aging outcomes such as walking and cognition. For more clinical trials on resveratrol, see clinicaltrials.gov.
Last Updated: 16 Oct 2015
Further Reading
No Available Further Reading
Overview
Name: PRX-03140
Synonyms: Potassium salt
Chemical Name: 7-isopropyl-6-oxo-5(3-piperidin-1-yl-propylcarbamoyl)-6,7-dihydro-thieno[2,3-b]pyridine-4-olate
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Inactive)
Company: Epix Pharmaceuticals
Last Updated: 17 Dec 2013
Further Reading
No Available Further Reading
Overview
Name: Propentofylline
Synonyms: HWA 285, PPF
Therapy Type: Small Molecule (timeline)
Target Type: Unknown
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Background
Propentofylline is a xanthine derivative that inhibits adenosine uptake and also inhibits phosphodiesterase. It exerts inhibitory activity on the proliferation of microglial cells, the formation of oxygen free radicals, and the release of inflammatory cytokines. [Cochrane Library]
Last Updated: 16 Dec 2013
Further Reading
No Available Further Reading
Overview
Name: Physostigmine Salicylate
Synonyms: Synapton
Therapy Type: Small Molecule (timeline)
Target Type: Cholinergic System (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Forest Laboratories, Inc.
Background
Physostigmine salicylate is a reversible acetylcholinesterase inhibitor that effectively increases the concentration of acetylcholine and thus the stimulation of both nicotinic and muscarinic receptors due to the increase in available acetylcholine at the synapse.
Last Updated: 12 Dec 2013
Further Reading
No Available Further Reading
Overview
Name: Phenserine
Therapy Type: Small Molecule (timeline)
Target Type: Cholinergic System (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Inactive)
Company: Annovis Bio, TorreyPines Therapeutics, Inc.
Findings
Phenserine’s development was stopped after it failed to improve cognition or clinical measures in two Phase 3 trials (Sep 2005 news).
Last Updated: 19 Jun 2020
Further Reading
No Available Further Reading
Overview
Name: Azeliragon
Synonyms: PF-04494700, TTP488
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline), Inflammation (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Pfizer, TransTech Pharma, Inc., vTv Therapeutics LLC
Background
Azeliragon is an oral, small-molecule inhibitor of RAGE, a cell-surface receptor of the immunoglobulin superfamily. Expressed on multiple cell types, RAGE binds advanced glycation end products (AGEs); these are modified forms of lipids and proteins that become glycated when exposed to sugars. AGEs form during normal aging and in higher amounts in patients with diabetes. When bound to their receptors, AGEs cause inflammation and oxidative damage. RAGE also binds Aβ (Yan et al., 1996), and has been reported to mediate toxic effects of Aβ oligomers in neurons (Mar 2008 news). RAGE is upregulated in astrocytes and microglia in the hippocampi of people with AD (Lue et al., 2005) and is thought to mediate amyloid transport into the brain.
The RAGE antagonist azeliragon blocks this interaction; hence the rationale is that it could provide a combined treatment effect across glial inflammatory and amyloid-related processes. In preclinical studies, the compound decreased brain Aβ load in transgenic mice and improved their performance on behavioral assays.
Findings
This compound was discovered by TransTech Pharma as TTP488 and licensed to Pfizer as PF-04494700. In 2005, Pfizer ran a 10-week Phase 2 trial in 67 people with mild to moderate AD. This trial compared two doses—15 mg for six days followed by daily dosing of 5 mg, and 60 mg for six days followed by daily dosing of 20 mg—against placebo for safety and tolerability. Both doses were reported to be safe and well-tolerated, with more people in the treatment groups completing the trial than people in the placebo group (Sabbagh et al., 2011).
In 2007, Pfizer and the National Institute on Aging jointly funded a larger, 18-month Phase 2 trial run via the Alzheimer's Disease Cooperative Study (ADCS). This trial recruited 399 people with mild to moderate AD and evaluated the same doses for safety and efficacy as measured by the ADAS-cog. The higher dose was dropped after a six-month interim analysis flagged both safety signals and faster deterioration. The low dose was halted before its intended conclusion following a futility analysis that indicated no benefit; however, follow-up examination conducted after treatment was suspended did suggest a possible belated clinical benefit for the low dose (Nov 2011 conference news; Galasko et al., 2014). A subgroup analysis reported numerical group differences in mild AD, favoring the low dose over placebo, and linked higher plasma levels of TTP488 to slower cognitive decline (Burstein et al., 2014; Burstein et al., 2018).
Pfizer terminated its work with TTP488 in 2011. Pfizer had previously been developing this compound for diabetic neuropathy but discontinued this program as well.
In March 2013, TransTech Pharma announced in a press release that it had received fast-track designation from the Food and Drug Administration for this compound in Alzheimer's disease, and in July it announced that it had met with the FDA and was planning a pivotal trial in patients with mild to moderate AD.
In May 2015, TransTech Pharma became vTv Therapeutics.
One month before the name change, a Phase 3 trial of azeliragon began; it ran at 115 study locations in North America, Australia, New Zealand, South Africa, the U.K., and Ireland. Called STEADFAST, this study compared an 18-month course of the 5 mg daily dose to placebo in 880 people who have a clinical diagnosis of mild probable Alzheimer's disease and an MRI consistent with this diagnosis. The trial did not use CSF or amyloid PET to ascertain that Alzheimer's underlies the clinical symptoms. The twin primary outcomes were the ADAS-cog and CDR-sum of boxes; secondary outcomes included MRI and FDG-PET, plasma Aβ levels, and neuropsychiatric, functional, and cognitive measures.
In April 2018, vTv announced that STEADFAST had failed to meet its co-primary endpoint (April 2018 news). The trial was set to run until June 2019 but was terminated in June 2018. A two-year, open-label extension study to evaluate continued safety parameters and progression slopes had been projected to enroll 640 STEADFAST completers and run through 2020; however, in June 2018, with 298 people enrolled, it was terminated as well.
At subsequent conferences, the investigators reported subgroup analyses claiming a benefit on both the ADAS-cog and CDR-sum of boxes in participants with Type 2 diabetes and elevated concentrations of acetylated hemoglobin (HBA1c), a RAGE ligand (Nov 2018 conference news, AD/PD slide set; AAIC 2019 abstract).
In June 2019, vTv began a Phase 2/3 trial of azeliragon in a planned 300 participants with a clinical diagnosis of mild Alzheimer’s disease and high HbA1c. In the first phase, approximately 100 volunteers were to receive 5 mg drug, once a day, or placebo, for six months; the primary outcome is change from baseline in ADAS-Cog. Results from this group were expected by the end of 2020. In a second phase, an additional 200 were to be randomized to treatment or placebo for 18 months, with the primary endpoints of ADAS-Cog and CDR-sum of boxes at the end of treatment. The study was being conducted at 31 sites in the U.S. and Canada, and slated to end in 2023. At the end of 2020, the company announced azeliragon had missed its endpoint of slowing decline on the ADAS-Cog in 43 participants after six months (Dec 2020 news). The trial was terminated, and development of azeliragon for Alzheimer’s stopped.
For all trials of azeliragon, see clinicaltrials.gov and clinicaltrials.gov.
Clinical Trial Timeline
- Phase 2/3
- Phase 3
- Study completed / Planned end date
- Planned end date unavailable
- Study aborted
Sponsor |
Clinical Trial |
2013 |
2014 |
2015 |
2016 |
2017 |
2018 |
2019 |
2020 |
2021 |
2022 |
2023 |
2024 |
2025 |
2026 |
2027 |
2028 |
2029 |
2030 |
2031 |
2032 |
2033 |
2034 |
vTv Therapeutics LLC |
NCT02080364 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
vTv Therapeutics LLC |
NCT03980730 |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Last Updated: 02 Feb 2021
Further Reading
No Available Further Reading
Overview
Name: PBT2
Synonyms: PBT-2
Chemical Name: Hydroxyquinoline
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline), Metals
Condition(s): Alzheimer's Disease, Huntington's Disease
U.S. FDA Status: Alzheimer's Disease (Inactive), Huntington's Disease (Inactive)
Company: Alterity Therapeutics
Background
PBT2 is a metal protein-attenuating compound (MPAC) being developed for treatment of both Alzheimer's and Huntington's diseases. It is an 8-hydroxyquinoline derivative that disrupts the interaction between metals and the Aβ peptide in the brain. The rationale is that as an MPAC, PBT2 prevents Aβ accumulation while also restoring copper and zinc ion homoeostasis in cells. According to scientists at Alterity Therapeutics, formerly Prana Biotechnology Ltd., increasing bioactive metal levels in the aging brain accelerates formation of amyloid plaques as well as neurotoxic oxidative processes. PBT2 translocates copper and zinc ions into the cell, reducing their extracellular levels and thereby reducing metal-mediated Aβ aggregation. It does so by acting as a copper and zinc ionophore rather than a chelator. PBT2 was reported to improve indicators of synaptic health such as spine density and synaptic protein levels in APP transgenic mice (Bush and Tanzi, 2008; Crouch et al., 2011; Adlard et al., 2011).
Two studies reported that copper drives aggregation of the mutant huntingtin protein and that PBT2 improved motor performance and extended the lifespan in a mouse model of HD (see Fox et al., 2007; Cherny et al., 2012).
PBT2 is taken as an oral capsule and crosses the blood-brain barrier. PBT2 is the second-generation compound with improved brain penetrance and pharmacokinetics to PBT1, which was discontinued because of formulation and safety issues.
Findings
In 2007, Prana conducted a Phase 2 trial at sites in Australia and Sweden. The 12-week trial enrolled 78 people with early Alzheimer's disease and compared 50 mg and 250 mg of PBT2 taken once daily to placebo on safety, as well as some fluid biomarker and cognitive and global function scales. PTB2 was reported to have been safe and well-tolerated, without serious side effects or withdrawals related to adverse events. The higher dose reportedly reduced Aβ42 levels in CSF, though not in plasma. There was no statistically significant improvement on the ADAS-Cog battery; however, a treatment benefit was seen in individual executive function tests (see March 2008 news story, Lannfelt et al., 2008). A subsequent post hoc analysis ranking the treatment responses reported that the findings of benefit were more likely with the higher dose than the lower dose or placebo (Faux et al., 2010 ). These studies called for larger trials to test PBT efficacy.
In 2011, Prana conducted a second Phase 2 study of 250 mg PBT2 once daily in 42 people with prodromal or mild AD. This trial used amyloid imaging as its primary outcome. On March 31, 2014, Prana reported top-line results stating that there was no significant difference between the treatment and placebo groups (see news story and extensive expert commentary). Results of this trial were published (Villemagne et al., 2017). The treated group showed a 3 percent decline in amyloid over one year, but there was no change in the placebo group. Amyloid levels stabilized during a one-year, open label extension. For more details on this trial, see Australian New Zealand clinical trials registry.
In 2012/2013, the Huntington Study Group ran Reach2HD, a Phase 2 trial, at 20 research sites in the U.S. and Australia. This trial compared a six-month course of 100 or 250 mg of PBT2 once daily to placebo in 109 patients with mild to mid-stage Huntington's disease. Prana disclosed top-line results on February 18, 2014. On the primary endpoint of safety and tolerability, Prana reported that five people dropped out of the trial and that PBT2 met safety and efficacy criteria for the remaining participants. Ten serious adverse events occurred, nine of them in the PBT2 groups. Prana claims that all serious adverse events were unrelated to PBT2 except for one person who reported a worsening of Huntington's symptoms after the treatment period. Of seven secondary outcomes, cognition was prespecified as the main efficacy variable based on the previous Phase 2 results in AD. An executive-function composite score comprising Category Fluency and the Trail Making Test Part B showed a trend toward improvement in people taking the higher dose, and statistical significance in people with mild HD. This this was due to an improvement compared with placebo for the high dose in the Trail Making Test, which was also the basis for the positive effect seen in the AD trial. No treatment effect was seen on any other outcome (for detail and expert commentary, see Feb 2014 news story). Results were published after peer review (Huntington Study Group Reach2HD Investigators, 2015).
In 2014, Phase 1 trials evaluating the relationship of food and caffeine to the pharmacokinetics of PBT2, and a bioavailability study, recruited a total of 44 healthy volunteers. Results are posted on clinicaltrials.gov.
In 2014 and 2015, PBT2 received Orphan Drug Designation for Huntington's disease from regulators in the U.S. and Europe. In February 2015, Prana announced that the U.S. FDA had placed a partial clinical hold, which limited the allowed dose to below the level the company considered to be clinically useful. In December 2016, the company disclosed that European regulators had recommended further animal work to establish whether neurotoxicity detected in dog studies was reversible (press release). At that time, Prana said they were looking for a collaborator on this project. No further development activity of PBT2 for neurodegeneration has been announced.
In 2019, Prana changed its name to Alterity Therapeutics.
For clinical trials of PBT2, see clinicaltrials.gov.
Last Updated: 16 Oct 2023
Further Reading
No Available Further Reading
Comments
No Available Comments
Make a Comment
To make a comment you must login or register.