Therapeutics

Edonerpic

Tools

Back to the Top

Overview

Name: Edonerpic
Synonyms: T-817 MA, T 817
Chemical Name: 1-{3-[2-(1-benzothiophen-5-yl)ethoxy] propyl}-3-azetidinol maleate
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline), Unknown
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Toyama Chemical Co., Ltd.

Background

T-817MA is an orally available neurotrophic agent being developed for the treatment of Alzheimer's disease by Toyama Chemical, a company of the Fujifilm Group. T-817MA reportedly activates sigma receptors. The compound has been reported in various cell-based and preclinical models to protect neurons against Aβ-induced neurotoxicity and memory deficits (e.g. Hirata et al., 2005Nguyen et al., 2007; Kimura et al., 2009). T-817MA appears to act by promoting neurite outgrowth and preserving synaptic plasticity in the cortex and hippocampus (Takamura et al., 2013). 

A treatment benefit has also been reported in sensorimotor gating, which is a biomarker for cognitive deficits in schizophrenia, as well as in models of hearing loss; however, no clinical development is ongoing in these indications (Seo et al., 2008; Uehara et al., 2012; Yamashita et al., 2008).

Edonerpic was reported to enhance cortical synaptic plasticity and motor function recovery after stroke, both in mice (Abe et al., 2018Pines et al., 2019). In Abe et al., edonerpic’s target was proposed to be collapsin response mediator protein 2 (CRMP2), a cytosolic phosphoprotein involved in neurite outgrowth, ion channel trafficking, and synaptic plasticity. However, other investigators dispute that edonerpic binds to CRMP2 (Moutal et al., 2019, reviewed in Takahashi 2019Khanna et al., 2020). The compound was subsequently reported to act via CRMP2 and the Arc protein to improve long-term neurological recovery in a mouse model of traumatic brain injury (Chen et al., 2022).

Findings

Phase 1 development for T-817MA began in 2005. From 2008 to 2011, a multicenter Phase 2a trial in North America compared a once-daily dose of 224 mg of T-817MA to placebo in 373 patients with mild to moderate Alzheimer's disease who are stable on donepezil. The primary outcome was cognitive function as measured by the ADAS-cog; secondary outcomes were safety and overall impression using the ADCS Global Clinical Impression of Change (CGIC) and ADCS Activities of Daily Living (ADL). In this trial, only 72.1 percent of patients on placebo, versus 61.6 percent on T-817MA, completed the trial, limiting conclusions that can be drawn from its results. Within these limitations, T-817MA appeared to show potential to slow cognitive and functional decline. Numerical differences on cognitive, clinical, and functional readouts favored treatment but fell short of statistical significance. Results of a neuropsychiatric measure favored placebo, also without statistical significance. A small imaging sub-study indicated trends favoring T-817MA, again not statistically significantly. Side effects included diarrhea, nausea, dizziness, and headache; two serious adverse events were considered possibly related to the study drug (Schneider et al., 2013).

In March 2014, the North American consortium Alzheimer's Disease Cooperative Study (ADCS) started another one-year, Phase 2 study of T-817MA called Noble. It compared 224 mg to 448 mg of T-817MA once daily and to placebo. It enrolled 484 patients with mild to moderate Alzheimer's disease who were already taking donepezil or rivastigmine, with or without memantine. This study used the ADAS-cog and ADCS-CGIC as primary outcomes, and safety as well as ADCS-ADL, FAQ, NPI, and MMSE as secondary outcomes. Unlike most ADCS trials, this study received no federal funding but was fully sponsored by Toyama. This trial was completed in May 2017.

On July 19, 2017, Fujifilm Corporation announced that the Noble trial had failed to reach its primary endpoint, and that there were no differences between the groups on secondary endpoints. According to the press release, exploratory analyses spotted biomarker signals in CSF p-tau levels and hippocampal volume, and post-hoc analyses hinted at a possible subgroup effect on cognition. Fujifilm announced its intention to continue developing T-817MA (see company press release). In November 2017, results presented at CTAD confirmed that Edonerpic had failed to outperform placebo on the primary or any of the secondary outcomes. Some biomarkers showed changes, but the changes did not all trend in the same direction. Diarrhea, vomiting, and weight loss were more common in the treatment groups than placebo, and more people in the treatment arms discontinued the trial. Dropouts in the placebo, low and high dose were 11.4, 29.5, and 24.1 percent, respectively (see Dec 2017 conference news).

The complete trial data has been published (Jul 2019 newsSchneider et al., 2019). Among biomarkers, CSF phospho-tau181 and total tau decreased in the high-dose group compared with placebo, while Aβ40 and Aβ42 did not change. Treatment caused no change in hippocampal volume, but ventricular volume increased in the high-dose group.

In February 2016, another Phase 1 trial started. It compared pharmacokinetic parameters of a single dose of T-817MA between otherwise healthy volunteers with mildly, moderately, or severely impaired liver function and people with normal liver function. This trial enrolled 36 people in Florida, and was completed in August 2016.

In April 2019, the company began a Phase 2 trial, enrolling 45 people in Japan to evaluate whether edonerpic improves rehabilitation of partial arm paralysis due to stroke.

A Phase 2 trial began in December 2019 to assess edonerpic’s effect on disease-related biomarkers in 200 people with mild cognitive impairment due to AD, or mild AD, whose clinical diagnosis was confirmed by CSF Aβ and tau abnormality. Participants took 224 mg edonerpic daily for one month, then 448 mg or placebo for an additional 17 months. The primary outcome was change in CSF pTau181 from baseline. Secondary outcomes included CSF total tau, pTau217, Aβ, NfL, neurogranin, and YKL; plasma NfL and Aβ, clinical dementia rating, brain volume, EEG measures, safety, and pharmacokinetics. The trial enrolled 221 participants at 31 sites in Europe. It failed to meet its primary endpoint, according to results presented at the October 2023 CTAD conference. Edonerpic did not change CSF ptau181 compared to placebo. Treatment did not affect most of the secondary endpoints, including the CDR-SB and the Cognitive-Functional Composite, and EEG measures. Just more than 80 percent of the treated group completed the trial. Treatment was deemed safe, although those on edonerpic ended with lower total brain volume and higher ventricular volume, with no change in hippocampal volume.

For details on T-817MA trials, see clinicaltrials.gov.

Last Updated: 06 Nov 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

References

Therapeutics Citations

  1. Donepezil
  2. Rivastigmine
  3. Memantine

News Citations

  1. At Least We Know These Don’t Work: Negative Trials at CTAD
  2. Epilogue on Edonerpic: It Does Not Work in Alzheimer’s

Paper Citations

  1. . The neuroprotective and neurotrophic agent T-817MA for Alzheimer’s disease: Randomized, double-blind, placebo-controlled proof-of-concept trial outcomes. Alzheimer's & Dementia, July 2013
  2. . Safety and Efficacy of Edonerpic Maleate for Patients With Mild to Moderate Alzheimer Disease: A Phase 2 Randomized Clinical Trial. JAMA Neurol. 2019 Jul 8; PubMed.
  3. . A novel neurotrophic agent, T-817MA [1-{3-[2-(1-benzothiophen-5-yl) ethoxy] propyl}-3-azetidinol maleate], attenuates amyloid-beta-induced neurotoxicity and promotes neurite outgrowth in rat cultured central nervous system neurons. J Pharmacol Exp Ther. 2005 Jul;314(1):252-9. PubMed.
  4. . Ameliorative effects of a neuroprotective agent, T-817MA, on place learning deficits induced by continuous infusion of amyloid-beta peptide (1-40) in rats. Hippocampus. 2007;17(6):443-55. PubMed.
  5. . T-817MA, a neurotrophic agent, ameliorates the deficits in adult neurogenesis and spatial memory in rats infused i.c.v. with amyloid-beta peptide. Br J Pharmacol. 2009 Jun;157(3):451-63. PubMed.
  6. . Effects of the neurotrophic agent T-817MA on oligomeric amyloid-β-induced deficits in long-term potentiation in the hippocampal CA1 subfield. Neurobiol Aging. 2013 Oct 7; PubMed.
  7. . T-817MA, a novel neurotrophic compound, ameliorates phencyclidine-induced disruption of sensorimotor gating. Psychopharmacology (Berl). 2008 Apr;197(3):457-64. PubMed.
  8. . T-817MA, a novel neurotrophic agent, ameliorates loss of GABAergic parvalbumin-positive neurons and sensorimotor gating deficits in rats transiently exposed to MK-801 in the neonatal period. J Psychiatr Res. 2012 May;46(5):622-9. Epub 2012 Feb 18 PubMed.
  9. . Neuroprotective effects of T-817MA against noise-induced hearing loss. Neurosci Res. 2008 May;61(1):38-42. PubMed.
  10. . CRMP2-binding compound, edonerpic maleate, accelerates motor function recovery from brain damage. Science. 2018 Apr 6;360(6384):50-57. PubMed.
  11. . Edonerpic Maleate: A Promising Pharmacological Agent for Stroke Recovery. Neurosurgery. 2019 Jan 1;84(1):E3-E4. PubMed.
  12. . Evaluation of edonerpic maleate as a CRMP2 inhibitor for pain relief. Channels (Austin). 2019 Dec;13(1):498-504. PubMed.
  13. . Novel synaptic plasticity enhancer drug to augment functional recovery with rehabilitation. Curr Opin Neurol. 2019 Dec;32(6):822-827. PubMed.
  14. . Druggability of CRMP2 for Neurodegenerative Diseases. ACS Chem Neurosci. 2020 Sep 2;11(17):2492-2505. Epub 2020 Aug 4 PubMed.
  15. . Edonerpic maleate regulates glutamate receptors through CRMP2- and Arc-mediated mechanisms in response to brain trauma. Cell Death Discov. 2022 Mar 4;8(1):95. PubMed.

External Citations

  1. Noble
  2. company press release
  3. Phase 2 trial
  4. clinicaltrials.gov

Further Reading

Papers

  1. . T-817MA, a novel neurotrophic agent, improves sodium nitroprusside-induced mitochondrial dysfunction in cortical neurons. Neurochem Int. 2006 Jan;48(2):124-30. PubMed.
  2. . A neuroprotective agent, T-817MA (1-{3-[2-(1-benzothiophen-5-yl)ethoxy]propyl} azetidin-3-ol maleate), prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice. Neuropharmacology. 2008 Oct;55(5):654-60. PubMed.
  3. . Blocking Effects of Human Tau on Squid Giant Synapse Transmission and Its Prevention by T-817 MA. Front Synaptic Neurosci. 2011;3:3. PubMed.
  4. . Protective effect of neurotrophic agent T-817MA against inner ear barotrauma in the guinea pig. J Pharmacol Sci. 2011;117(1):67-70. Epub 2011 Aug 25 PubMed.
  5. . T-817MA, but Not Haloperidol and Risperidone, Restores Parvalbumin-Positive γ -Aminobutyric Acid Neurons in the Prefrontal Cortex and Hippocampus of Rats Transiently Exposed to MK-801 at the Neonatal Period. ISRN Psychiatry. 2012;2012:947149. Print 2012 PubMed.
  6. . A systematic review on drugs for synaptic plasticity in the treatment of dementia. Ageing Res Rev. 2022 Nov;81:101726. Epub 2022 Aug 27 PubMed.