Therapeutics

ASN51

Tools

Back to the Top

Overview

Name: ASN51
Synonyms: ASN121151
Therapy Type: Small Molecule (timeline)
Target Type: Tau (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 1)
Company: Asceneuron SA

Background

ASN51 is a small-molecule inhibitor of O-GlycNAcase (OGA), the glycoside hydrolase enzyme that removes O-linked N-acetylglucosamine (N-GlcNAc) from proteins. Asceneuron is developing ASN51 as a potential treatment for Alzheimer’s and Parkinson’s diseases. It is a second-generation version of its lead OGH inhibitor, ASN120290, which is intended to treat the rare tauopathy progressive supranuclear palsy. ASN51 is taken in capsule form.

The rationale for targeting OGA originated in the observation that addition of N-GlcNAc to tau reduces its propensity to form toxic aggregates (Gong et al., 2005; Liu et al., 2004). OGA inhibitors promote glycosylation, prevent aggregation, and appear to stabilize tau in a soluble, nonpathogenic form. O-GlcNAc modification was reported to reduce the aggregation and toxicity of the α-synuclein protein as well (Marotta et al., 2015Levine et al., 2019). This modification appears to promote formation of an α-synuclein fibril strain that does not propagate and is not neurotoxic in vivo (Balana et al., 2024).Thus, OGA inhibitors could potentially slow or prevent the progression of neurodegeneration due to these proteins.

No preclinical work is published on ASN51. However, Asceneuron has published data on ASN90, their first-generation OGA inhibitor in development for the primary tauopathy progressive supranuclear palsy. In the P301S mouse model of tauopathy, daily oral dosing enhanced brain tau glycosylation, prevented the development of tau tangles, improved motor behavior and breathing, and increased survival (Permanne et al., 2022). In this study, tau glycosylation in peripheral blood cells mirrored modification in the CNS, and thus was suggested as a potential pharmacodynamic marker of OGA inhibition for clinical trials. Also in this work, ASN90 was shown to enhance α-synuclein glycosylation, and slow the progression of motor impairment in a transgenic model of Parkinson’s disease.

A different OGA inhibitor, thiamet G, has also been reported to increase glycosylated tau, reduce tau neurofibrillary tangle numbers, and decrease neuronal cell loss in three different mouse strains expressing P301L mutant human tau (Yuzwa et al., 2012; Graham et al., 2014; Hastings et al., 2017). One lab found that thiamet G treatment resulted in better motor skills, higher body weight, and longer lifespan (Borghgraef et al., 2013). Thiamet G or OGA knockdown was also shown to reduce the uptake of synuclein fibrils by cells (Tavassoly et al., 2020). Fibril uptake is a proposed mechanism for the spread of α-synuclein pathology in the brain.

Findings

In June 2021, Asceneuron began a Phase 1 first-in-human trial in Melbourne, Australia, investigating the safety, tolerability, pharmacokinetics, and pharmacodynamics of ASN51. The placebo-controlled study planned to test single and multiple doses of oral capsules in 64 healthy adults, followed by a 10-day evaluation in 12 people with AD. This study was terminated in August 2022 after completion of the single-dosing portion, and one multiple-dose cohort. The stated reasons were site delays and the impact of COVID.

The company presented results from healthy adults at the December 2022 CTAD conference. Single doses of 20 and 50 mg in young adults and multiple 20 mg doses in eight older adults were safe, with no serious or severe adverse events. The most common side effects were headache and musculoskeletal ache. One person had moderate, transient dizziness. ASN51 produced predictable, dose-proportional plasma PK with a half-life of 40-50 hours, that was unaffected by food or age. CSF concentrations were in the active range, and highly correlated with plasma levels, suggesting the drug freely diffused into the CNS. A pharmacodynamic assay found tau glycosylation to increase in peripheral blood cells after a single 20 or 50 mg dose.

A PET study with an OGA ligand revealed significant target occupancy for up to two days after a single dose of 5 mg or higher. Based on PET results, the investigators estimated that a 10 mg dose would sustain greater than 95 percent brain OGA occupancy for 24 hours.

From February to June of 2023, the company conducted a pharmacokinetic and pharmacodynamic study after repeated dosing in 12 healthy men. According to a presentation at the October 2023 CTAD, 14 days treatment with 10 or 20 mg daily produced no serious adverse events or safety signs, and no study withdrawals. Plasma levels were dose-proportional, reached steady-state by one week, and were maintained for up to a week after dosing stopped. PET revealed greater than 90 percent brain OGA occupancy within hours after the first dose, and three days after the last dose. Two weeks of drug elevated tau glycosylation in peripheral blood cells. Food did not affect this pharmacodynamic marker.

The company is planning a Phase 2 study, to start in 2024.

In February 2025, a study began to assess drug-drug interactions with inhibitors of three cytochrome p450 isotypes. The study will test, in 48 healthy adults, the effect of simultaneous dosing with fluvoxamine, itraconazole, or paroxetine on the pharmacokinetics of ASN51. It will finish in May 2024.

For details on ASN51 trials, see clinicaltrials.gov.

Last Updated: 07 Mar 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

References

Paper Citations

  1. . Post-translational modifications of tau protein in Alzheimer's disease. J Neural Transm. 2005 Jun;112(6):813-38. PubMed.
  2. . O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease. Proc Natl Acad Sci U S A. 2004 Jul 20;101(29):10804-9. PubMed.
  3. . O-GlcNAc modification blocks the aggregation and toxicity of the protein α-synuclein associated with Parkinson's disease. Nat Chem. 2015 Nov;7(11):913-20. Epub 2015 Oct 12 PubMed.
  4. . α-Synuclein O-GlcNAcylation alters aggregation and toxicity, revealing certain residues as potential inhibitors of Parkinson's disease. Proc Natl Acad Sci U S A. 2019 Jan 29;116(5):1511-1519. Epub 2019 Jan 16 PubMed.
  5. . O-GlcNAc forces an α-synuclein amyloid strain with notably diminished seeding and pathology. Nat Chem Biol. 2024 Feb 12; PubMed. Correction.
  6. . O-GlcNAcase Inhibitor ASN90 is a Multimodal Drug Candidate for Tau and α-Synuclein Proteinopathies. ACS Chem Neurosci. 2022 Apr 20;13(8):1296-1314. Epub 2022 Mar 31 PubMed.
  7. . Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol. 2012 Apr;8(4):393-9. PubMed.
  8. . Increased O-GlcNAcylation reduces pathological tau without affecting its normal phosphorylation in a mouse model of tauopathy. Neuropharmacology. 2014 Apr;79:307-13. Epub 2013 Dec 8 PubMed.
  9. . Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener. 2017 May 18;12(1):39. PubMed.
  10. . Increasing brain protein O-GlcNAc-ylation mitigates breathing defects and mortality of Tau.P301L mice. PLoS One. 2013;8(12):e84442. Epub 2013 Dec 23 PubMed.
  11. . Pharmacological inhibition and knockdown of O-GlcNAcase reduces cellular internalization of α-synuclein preformed fibrils. FEBS J. 2020 May 4; PubMed.

Other Citations

  1. ASN120290

External Citations

  1. clinicaltrials.gov

Further Reading

No Available Further Reading