Therapeutics

Zagotenemab

Tools

Back to the Top

Overview

Name: Zagotenemab
Synonyms: LY3303560
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Tau (timeline), Unknown
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Eli Lilly & Co.

Background

Zagotenemab is a humanized anti-tau antibody derived from MCI-1, Peter Davies' mouse monoclonal antibody against an early pathological conformation of tau. Zagotenemab binds and neutralizes soluble tau aggregates.

Preclinical studies in tau transgenic mice showed that immunotherapy with MCI-1 reduced levels of hyperphosphorylated insoluble tau levels and neurofibrillary pathology (Chai et al., 2011).

At the 2017 AAIC meeting, Lilly presented data from surface plasmon resonance binding and ELISA assays to assess LY3303560's selectivity to aggregates over monomer and characterize its epitope. LY3303560 reportedly had high affinity to soluble tau aggregate in vitro, at a KD of below 220 picomolar, compared to monomer, KD of 235 nanomolar. The antibody reportedly recognizes a conformational epitope whose primary epitope is in tau's N-terminal region. Intravenous administration to monkeys indicated clearance of 0.15 ml/h/kg and a half-life of 13 days. SC administration indicated a bioavailability of 79 percent, and rat CSF concentration was 0.1 percent of plasma at 24 hours after IV administration (Alam et al., 2017).

Findings

From April 2016 to July 2018, Lilly ran a first-in-human trial of LY3303560 in 110 people, both healthy volunteers and people whose MCI due to AD or mild to moderate AD was ascertained with a positive amyloid PET scan. This study evaluated a single, escalating intravenous infusion or subcutaneous injection of LY3303560 or placebo. It measured adverse effects up to 85 days after this dose, as well as exposure and maximal achieved drug concentration in both serum and CSF. This study required a four-day stay in a clinical research unit and 10 follow-up visits. It was conducted in California and Maryland. Results are posted on clinicaltrials.gov.

From January 2017 to June 2019, a second Phase 1 study intravenously delivered multiple escalating doses of LY3303560 or placebo to 24 people with the same diagnosis. Over a planned treatment period of six months plus four months of follow-up, the trial measured adverse events and pharmacokinetic parameters. Notably, this trial infused LY3303560 or placebo, and amyloid and tau PET tracers, in the same session. The study initially was going to measure change in tau PET as a secondary outcome, but dropped this measure in February 2018. This trial ran at 12 sites in the U.S., the U.K., and Japan. According to published results, 22 patients were randomized to 70 or 210 mg antibody, or placebo, for up to 49 weeks. The study was stopped before running two planned higher dose cohorts of 700 and 1,400 mg, because the investigators realized they would likely not reach an effective dose. No dose-limiting adverse events occurred. Pharmacokinetics were linear, and typical for a monoclonal antibody. Treatment caused a dose-dependent increase in plasma tau; tau-PET was not changed (Willis et al., 2023).

In April 2018, Phase 2 began with a first efficacy trial enrolled 360 people who had a gradual and progressive decline in memory for at least six months. The study compared 1,400 mg or 5,600 mg monthly intravenous doses to placebo. Change from baseline on Lilly's integrated Alzheimer's Disease Rating Scale (iADRS) after two years was the primary outcome; secondary measures included ADAS-Cog13, ADCS-iADL, CDR-SB, MMSE, the CogState Brief Battery (CBB), as well as tau PET, volumetric MRI, the Columbia Suicide Severity Rating Scale (C-SSRS), and antigenicity of LY3303560, also out to two years. This trial ran at 60 sites in North America and Japan. It finished enrolling in August 2019 and ran until August 2021. In an October 2021 investor call, Lilly disclosed that the trial had missed its primary endpoint, and the company was ending development of zagotenemab (slides 5, 17 in presentation). Full trial results were published after peer review (Fleisher et al., 2024).

For all trials, see clinicaltrials.gov.

Clinical Trial Timeline

  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035
Eli Lilly & Co. NCT03518073
N=285

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

PF-06751979

Tools

Back to the Top

Overview

Name: PF-06751979
Synonyms: BACE inhibitor
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Pfizer

Background

Pfizer developed the BACE 1 inhibitor PF-06751979 for the treatment of Alzheimer's disease to Phase 1. This inhibitor is selective for BACE1 over BACE2 and the related aspartyl protease Cathepsin D. It enters the brain, and reduces the CSF Aβ42 concentration in mice and dogs. In contrast to nonselective BACE inhibitors, PF-06751979  induced no depigmentation in human melanocyte cultures, and chronic administration to dogs for up to nine months causes no loss in coat pigmentation (O’Neill et al., 2018). This drug was given as an oral suspension.

Findings

Between July 2015 and July 2016, Pfizer conducted a first-in-human, single- and multiple-ascending-dose trial of PF-06751979 in 55 healthy people in California. Doses ranged from 3 to 160 mg; treatment duration was up to two weeks. Primary outcome measures included a wide range of safety measures; secondary outcomes included plasma pharmacokinetics, as well as quantification of various CSF Aβ fragments at baseline and at two weeks.

In June 2016, a second trial started enrolling 46 elderly volunteers to evaluate single- and multiple-ascending doses ranging from 200 to 700 mg against placebo. Outcomes included numerous safety and pharmacokinetic and -dynamic measures, as well as measurement of these Aβ fragments: Aβ1-40, Aβ1-42, Aβ total, Aβx-40, Aβx 42, sAPPα, sAPPβ. This trial took place in Belgium. At the 2017 AAIC conference, Pfizer published results of these two trials as showing PF-06751979 to have been safe and well-tolerated, and to have reduced plasma and CSF Aβ in a dose-dependent fashion (podium presentation by Qiu et al., 2017).

In April 2017, a third trial evaluated the effect of this drug on the pharmacokinetics of midalozam in 12 healthy volunteers.P fizer published results of all three trials as showing PF-06751979 to have been safe and well-tolerated, with no drug interaction with midalozam. PF-06751979 reduced plasma and CSF Aβ in a dose-dependent fashion (Qui et al., 2019).

In January 2018, Pfizer announced it was ending its research and development in neurology, including this compound.

For all trials on this compound, see clinicaltrials.gov.

Last Updated: 29 Nov 2019

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

BIIB118

Tools

Back to the Top

Overview

Name: BIIB118
Synonyms: PF-05251749
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease, Parkinson's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 1), Parkinson's Disease (Phase 1)
Company: Biogen

Background

PF-05251749 is an inhibitor of Casein kinase 1 delta and epsilon (CK1δ and ε). It was being developed by Pfizer for the treatment of Alzheimer's disease, and is now owned by Biogen. It is taken by mouth.

CK1δ and ε are key regulators of the circadian clock, and are upregulated in AD brain. Early in AD, disruption of circadian cycles, including sleep, is thought to contribute to the accumulation of amyloid, and subsequent cognitive decline. Later in disease, changes in circadian and sleep cycles lead to abnormal late-day and nighttime activity, a major reason why people go into nursing homes.

Besides affecting circadian cycling, CK1δ and ε phosphorylate tau and are found in neurofibrillary tangles (Yasojima et al., 2000; Chen et al., 2017; Gu et al., 2019). CK1δ and ε also localize to granulovacuolar degeneration bodies, pathological protein assemblies found in the brains of people with AD and other neurodegenerative diseases (Wiersma et al., 2019; Riku et al., 2019).

No preclinical data has been published on PF-05251749; however, work with the Pfizer CK1 δ and ε dual inhibitor PF-670462 has been reported in two AD mouse models. In APP/PS1 mice, treatment with the inhibitor improved circadian rhythmicity and cognitive performance, while reducing brain amyloid (Sundaram et al., 2019). In 3X-Tg-AD mice, PF-670462 normalized alterations in hippocampal AD and clock-related pathways, including changes in proteins involved in synaptic plasticity and amyloid precursor protein processing. The compound also reduced working memory deficits and normalized circadian behaviors (Adler et al., 2019).

Findings

In 2015, Pfizer conducted a single-site, single-ascending-dose study in 32 healthy volunteers, assessing its propensity to induce movement disorders and affect mood and fatigue, as well as pharmacokinetics and exposure in CSF.

In March 2016, the company started an ascending-multiple-dose study of 50 to 900 mg daily of this compound in 97 healthy elderly volunteers at two sites in Florida. This trial also assessed suicidality and melatonin production, and expanded the number of pharmacokinetic measures. One arm administered PF-05251749 together with melatonin.

In January 2018, Pfizer announced it was ending its neurology research and development, including this compound.

In January 2020, Biogen acquired PF-05251749 (press release). The company announced plans to develop the drug for the treatment of sundowning in Alzheimer’s and irregular sleep-wake rhythm disorder in Parkinson’s, starting with a Phase 1b study in late 2020.

For all trials of this drug, see clinicaltrials.gov

Last Updated: 11 Sep 2020

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

LY2599666

Tools

Back to the Top

Overview

Name: LY2599666
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Eli Lilly & Co.

Background

LY2599666 is an Fc-less, antigen-binding fragment of a monoclonal anti-Aβ antibody linked to polyethylene glycol. It targets soluble Aβ monomer and is delivered by subcutaneous injection. 

Findings

In December 2015, Eli Lilly started a large Phase 1 trial, comparing LY2599666 to placebo and to solanezumab, the company's anti-Aβ antibody, in 130 volunteers. Solanezumab was administered by intravenous infusion, whereas LY2599666 was injected under the skin. This multicenter study in the United States and Japan compared these two agents on the same safety, pharmacokinetic, and plasma Aβ outcomes. The trial enrolled people who were either cognitively healthy or had a clinical diagnosis of amnestic MCI due to AD or mild to moderate Alzheimer's ascertained by amyloid PET. The trial compared a single injection of LY2599666, a three-month course of weekly injections of up to 200 mgs LY2599666, to a three-month course of either weekly or monthly infusions of solanezumab. In January 2017, Lilly terminated this compound, and at AAIC in July of 2017, reported that it was generally well tolerated but had low target engagement (Hake et al, 2017).

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

Therapeutics

LY3202626

Tools

Back to the Top

Overview

Name: LY3202626
Synonyms: BACE Inhibitor
Therapy Type: Small Molecule (timeline)
Target Type: Amyloid-Related (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Eli Lilly & Co.

Background

LY3202626 is a potent small-molecule inhibitor of BACE1, the β-secretase enzyme that cleaves the APP protein to release its C99 fragment. C99 gives rise to various species of Aβ peptide during its subsequent cleavage by γ-secretase. The rationale of BACE inhibition is that it represents an upstream interference with the amyloid cascade. This treatment approach is sometimes envisioned as long-term maintenance therapy to limit Aβ production after an initial round of immunotherapy to remove existing amyloid deposits. 

The potential advantage of LY3202626 over earlier compounds is its high potency. At the 2016 AAIC conference, Lilly reported that 1 mg/day of LY3202626 reduced CSF Aβ40 by half after two weeks (Aug 2016 conference news). It is formulated in capsule form.

Findings

Between December 2014 and February 216, Eli Lilly conducted a Phase 1 trial in four sequential parts. Parts A to C enrolled healthy people, part D enrolled people with AD, for a combined 136 participants. In part A, participants received placebo or 0.1 to 45 mg of the drug as a single dose and were monitored for safety. In part B, volunteers received 1.6 to 26 mg or placebo as a single dose, and serial plasma and CSF samples were taken for pharmacokinetic and pharmacodynamic analysis. Part C entailed multiple dosing up to 26 mg or placebo for two weeks. Part D administered 6 mg/day for two weeks. CSF was sampled at baseline in parts C and D, and again 24 hours after the final dose. At AAIC 2016, Lilly reported that LY3202626 was well-tolerated at all doses, without adverse events up to 42 days after the last dose. The compound’s half-life in plasma was 21 hours; it readily crossed the blood-brain barrier and dose-dependently reduced Aβ40 and Aβ42 in plasma and CSF. At 1, 6, and 26 mg/day over two weeks, LY3202626 reduced Aβ40 in CSF by 50, 75, and 90 percent, respectively (Aug 2016 conference newsWillis et al. 2016).

In September 2015, a second Phase 1 trial evaluated pharmacokinetic parameters of labeled LY3202626 in eight healthy volunteers, and from January to April 2017, a third Phase 1 trial in 30 healthy volunteers compared two different formulations and evaluated food effects on the pharmacokinetics of LY3202626.

In June 2016, Lilly started a Phase 2 trial called NAVIGATE-AD. Initially listed as aiming for 500 participants, this trial was cut back to 380 in August 2017, and terminated following an interim analysis in August 2018, when enrollment stood at 316. Enrollees had mild AD dementia as diagnosed by NIA/AA criteria, a positive florbetapir scan, and an MMSE between 20 and 26. They took either of two doses of LY3202626 or placebo once daily for a year, and were evaluated primarily with tau PET for their neurofibrillar pathology burden at baseline and one year. Secondary outcomes included safety measures of ARIA, pharmacokinetics of drug and Aβ in blood—but not CSF—as well as various efficacy measures such as the ADAS-cog13, the ADCS-iADL, and the integrated Alzheimer's Disease rating scale (iADRS). The trial was to take place at 76 centers in North America, Japan, and Australia, and to run through summer 2018. At the 2018 CTAD conference, Lilly reported that participants in this trial who had the longest exposure to LY3202626 showed hints of a cognitive deficit on the MMSE and ADAS-Cog13 (Nov 2018 conference news). Trial results have been published (Lo et al., 2021). The full analysis revealed no differences in tau PET, amyloid deposition, or measures of cognition and function in treated groups compared to placebo. Treatment was associated with brain volume loss in the hippocampus and other regions. A significant increase in psychiatric adverse events was noted for both doses compared to placebo, consistent with findings from trials of other BACE inhibitors, e.g. verubecestat.

In December 2017, Lilly began TRAILBLAZER, a combination trial of two investigational drugs targeting different points in the amyloid cascade. This Phase 2 was to evaluate safety, tolerability, and efficacy of an 18-month course of this BACE inhibitor in combination with donanemab, a monoclonal antibody against a pyroglutamate form of Aβ aggregated in plaques. It was to enroll 375 participants whose memory had been worsening for at least six months, and who met a cutoff on the CogState Brief Battery and had a positive tau PET scan. Because the antibody requires an infusion and the BACE inhibitor comes as a capsule, the three treatment arms in this blinded, placebo-controlled trial were to be as follows: One group to receive intravenous antibody plus placebo administered orally; the second group to receive intravenous antibody plus BACE inhibitor orally; the third group to receive both an intravenous and an oral placebo. Hence the trial was to evaluate the antibody alone and the antibody in combination with this BACE inhibitor, but not LY3202626 alone. The primary outcome is change on the Integrated Alzheimer's Disease Rating Scale (iADRS), a combined cognitive/functional measure for early stage AD developed by Lilly (Wessels et al., 2015). Secondary measures include the ADAS-Cog13, CDR-Sum of Boxes, MMSE, ADCS-iADL, as well as amyloid and tau PET and volumetric MRI. To ease the logistical burden of trial participation, this study offers rides to and from study sites via a partnership between GAP and the ridesharing company Lyft (press release). The study takes place at 69 sites in North America and is set to run until March 2020.

In October 2018, Lilly discontinued the BACE inhibitor arm of this trial; evaluation of the antibody remains ongoing, with a revised enrollment estimate of 266 participants. 

For all trials on this drug, see clinicaltrials.gov.

Clinical Trial Timeline

  • Phase 2
  • Study completed / Planned end date
  • Planned end date unavailable
  • Study aborted
Sponsor Clinical Trial 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035
Eli Lilly & Co. NCT02791191
N=316

Last Updated: 30 Jul 2021

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