Therapeutics

Tilavonemab

Tools

Back to the Top

Overview

Name: Tilavonemab
Synonyms: ABBV-8E12, C2N 8E12, HJ8.5
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Tau (timeline)
Condition(s): Progressive Supranuclear Palsy, Alzheimer's Disease
U.S. FDA Status: Progressive Supranuclear Palsy (Discontinued), Alzheimer's Disease (Discontinued)
Company: AbbVie, C2N Diagnostics, LLC

Background

C2N 8E12 is a humanized IgG4 antibody that was being developed by C2N Diagnostics and AbbVie to treat tauopathies. It recognizes an aggregated, extracellular form of pathological tau, binding to tau's N-terminus. This form of tau has been implicated in transneuronal propagation of tau pathology in cell-based and mouse models, and postulated to explain the stereotypical progression of tau pathology in Alzheimer's disease (Clavaguera et al., 2009Braak and Del Tredici, 2011). 

8E12 differs from some other anti-tau antibodies in that its mechanism of action requires no uptake into neurons. The mouse version of this antibody reportedly blocked seeding in a cell-based tau sensor assay and uptake of AD-derived tau by primary neurons (Kfoury et al., 2012Funk et al., 2015). In P301S tau-transgenic mice, it was reported to reduce brain neurofibrillary pathology, insoluble tau, microgliosis, seeding activity by the lysate of treated brain, brain atrophy, and deficits in the conditioned fear response (Yanamandra et al., 2013Yanamandra et al., 2015).

In July 2015 and April 2016, the FDA and EMA, respectively, granted this antibody orphan-drug designation for the treatment of progressive supranuclear palsy, a tauopathy affecting about 20,000 people in the United States, and 33,000 in Europe.

Findings

Between July 2015 and August 2016, AbbVie and C2N conducted a single-ascending-dose study at 12 centers in the United States. It compared four doses from 2.5 to 50 mg/kg of 8E12 to placebo in successive, three-to-one randomization groups and followed them to 84 days after dosing. The 30 participants met modified NINDS-SPSP criteria, and their MRI scans were consistent with a PSP diagnosis. The goal was to find the maximum tolerated dose; outcomes included safety, tolerability, immunogenicity, and pharmacokinetics. At conferences in 2016 and 2017, results were presented as showing 8E12 to have been safe; a maximum tolerated dose was not determined in this trial. The antibody had a half-life of 27 to 37 days, with dose-related exposure in blood and a CSF-to-blood ratio of 0.18 to 0.38 percent (Aug 2017 conference newsWest et al., 2017). In April 2018, an open-label extension study started to assess 8E12's longer-term safety and tolerability in participants in this trial who are ineligible for its Phase 2 trial below. 

In October 2016, AbbVie started a Phase 2 trial in 400 people who met diagnostic criteria for Alzheimer's disease. Their early disease stage was defined by a CDR rating of 0.5, an MMSE of 22 or higher, and an RBANS of 85 or lower; AD is ascertained by a positive amyloid PET scan. This trial compared three doses of 8E12 to placebo, to be infused over a period of 96 weeks plus 16-week followup. The primary outcomes were decline on the CDR Sum of Boxes and adverse events. Besides blood-based pharmacokinetic parameters for the 8E12 antibody, secondary outcomes included a range of clinical and functional measures such as the ADAS-Cog14, RBANS, FAQ, ADCS-MCI-ADL-24. No tau-based target engagement outcomes were listed. The trial was conducted at 68 sites in North America, six European countries, Australia, and New Zealand, and was set to run until 2021. In March 2019, the trial stopped enrolling at 453 participants, and ended in July 2021. People who completed the study were offered a long-term safety extension of up to 5.5 years of dosing. This extension also ended in July 2021. According to results presented at the 2021 CTAD conference, tilavonemab did not change the decline on the CDR-SB, or any of the cognitive for functional secondary outcomes, nor did it lower brain atrophy or levels of plasma neurofilament light (Nov 2021 conference news). Results were published after peer review (Florian et al., 2023). An autopsy of one patient who had received 2,000 mg antibody per month for 14 months revealed typical PSP pathology, with no pathological modifications attributed to treatment (Beck et al., 2023).

In December 2016, AbbVie started a Phase 2 PSP efficacy study comparing 2,000 or 4,000 mg of tilavonemab to placebo infused for one year. It enrolled 378 people aged 40 or older who had had PSP symptoms for fewer than five years. Primary outcomes were decline on the PSP Rating Scale (PSPRS) Total Score and adverse events; secondary outcomes included pharmacokinetic, MRI, as well as global and Parkinson’s clinical measures. The trial was conducted at 66 sites in North America, Australia, France, Germany, Italy, Spain, and Japan; it was set to run until June 2020. In January 2018, AbbVie and C2N started a four-year extension in people who completed the placebo-controlled phase; they were to receive ABBV-8E12 but stay randomized to dose. Primary outcomes were decline on the PSPRS Total Score for up to five years; secondary outcomes included change on Parkinson’s and global clinical measures.

In July 2019, an interim analysis of the Phase 2 PSP trial data found no benefit of antibody over placebo, and AbbVie stopped the study and the ongoing open-label extension (Jul 2019 news). The company also discontinued preapproval access for people with primary tauopathies. Trial results were subsequently published. Target engagement was confirmed by a decrease in CSF free tau and higher plasma total tau in treated groups, but the antibody showed no efficacy. No treatment-related adverse events were reported (Höglinger et al., 2021; Koga et al., 2021Höglinger, 2021Grossmann, 2021). More negative results of N-terminal anti-tau antibodies have been reported, as well (Mar 2021 conference news).

In July, 2021, AbbVie announced it would stop development of tilavonemab and delisted it from its pipeline.

For details, 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
AbbVie NCT02880956
N=

Last Updated: 07 Feb 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

S-Equol

Tools

Back to the Top

Overview

Name: S-Equol
Synonyms: Aus-131
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 1)
Company: Ausio Pharmaceuticals, LLC

Background

S-Equol is a selective agonist of the estrogen receptor-β, which is expressed on mitochondria. This compound is not a hormone, but rather the S-enantiomeric metabolite of daidzein, a soy-based isoflavone (Setchell et al., 2005). In humans and other mammals, including horses, S-Equol is normally made by intestinal bacteria. This synthesized formulation Aus-131 was originally developed for the treatment of menopausal symptoms and hyperplasia of the prostate.

Unlike estrogen receptor-α, which is expressed in the uterus and breast, ER-β is more strongly expressed in the brain. Estrogen receptor-β is considered a better target for neuroprotection, and agonists of this receptor subtype (SERMs) have been reported to improve hippocampal synaptic function and memory in mice (Tiwari-Woodruff et al., 2007Feb 2008 news). In cultured rat hippocampal neurons and a mouse ovariectomy model, S-Equol was reported to potentiate mitochondrial function (Yao et al., 2013).

Findings

In July 2015, a Phase 1 study at the University of Kansas Medical Center began enrolling 15 women with very mild or mild AD for a placebo-controlled trial of a six-week course of a twice-daily, 10 mg S-Equol pill or placebo. The primary outcome is activity of the platelet mitochondrial COX enzyme as an indicator of mitochondrial function. 

A separate formulation of three phytoSERMS—S-Equol, daidzein, and genistein—is being evaluated clinically, as well (Dec 2014 conference newsclincialtrials.gov).

For all trials of Aus-131, see clinicaltrials.gov.

Last Updated: 16 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

Neflamapimod

Tools

Back to the Top

Overview

Name: Neflamapimod
Synonyms: VX-745
Therapy Type: Small Molecule (timeline)
Target Type: Inflammation (timeline)
Condition(s): Alzheimer's Disease, Dementia with Lewy Bodies, Huntington's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 2), Dementia with Lewy Bodies (Phase 2), Huntington's Disease (Discontinued)
Company: CervoMed, Inc.

Background

Neflamapimod selectively inhibits the alpha isoform of the mitogen-activated serine/threonine protein kinase p38 MAPK, a "stress kinase" considered a drug target in CNS diseases (Duffy et al., 2011). Previously called VX-745, neflamapimod has been repurposed for evaluation in neurodegenerative diseases. VX-745 was discovered at Vertex Pharmaceuticals. It was being tested for rheumatoid arthritis, but was later discontinued, in part due to adverse effects in the central nervous system in preclinical studies (Haddad, 2001). This drug has been reported to reach higher concentrations in the CNS than peripheral blood.

p38 MAPKα is expressed in microglia and neurons. In microglia, the enzyme stimulates release of pro-inflammatory cytokines such as TNFα and IL-1β in response to a variety of stressors including Aβ42; in neurons, signaling via p38 MAPKα has been implicated in tau localization and neuronal plasticity (Yasuda et al., 2011Bachstetter et al., 2011Corrêa and Eales, 2012). In addition, p38 MAPKα regulates Ras-related protein Rab5, a key regulator of early endosomes whose role in endolysosomal and synaptic vesicle function has made it a target in neurodegenerative disease drug development (Germann and Alam, 2020). Other p38 MAPKα inhibitors are reported to slow progression in AD mouse models (Roy et al., 2015). 

In animal models, VX-745 reportedly shifts microglial activation from a pro-inflammatory to a phagocytic state, improving mitochondrial function, synaptic transmission, and memory. In 2-year-old Tg2576 mice treated with 3 mg/kg for two weeks, amyloid plaque load was halved. In aged rats, which develop more neuroinflammation than Tg2576 mice, VX-745 lowered IL-1β, increased the postsynaptic marker PSD95, and improved performance in the Morris water maze, though these effects were apparent at different doses (Dec 2014 conference news; Alam, 2015). In a rat ischemic stroke model, subacute treatment for six weeks dose-dependently promoted behavior, sensory and motor function recovery, and raised brain-derived neurotrophic factor (BDNF) protein levels (Alam et al., 2020). This drug also induces blood vessel dilation, and inhibits vascular inflammation in rats (Pandey et al., 2022Menon et al., 2023).

Recently, neflamapimod was reported to improve cholinergic dysfunction by inhibiting p38MAPK-driven Rab5 activity in neurons. In a mouse model of Rab5-dependent lysosomal pathology with cholinergic degeneration, neflamapimod reduced Rab5 activation, normalized the function of Rab5+ lysosomes, prevented degeneration of basal forebrain cholinergic neurons, and improved behavior in novel object recognition and open field tests (Jiang et al., 2022).  Basal forebrain cholinergic neurons are the main source of acetylcholine in the brain, and in people, their loss leads to cognitive decline and movement problems.

Neflamapimod is one of two p38 MAPKα inhibitors in development for AD. See also MW150.

Findings

In April 2015, two small, open-label Phase 2 trials began. Run by Parexel in Glendale, California, a pharmacology study compared two doses of VX-745, 40 mg or 125 mg, given orally for six weeks to 16 people with clinical diagnoses of MCI due to AD or mild AD, and brain hypometabolism as per FDG-PET scan. Primary outcomes were change on CSF biomarkers and FDG-PET; secondary outcomes were safety and exposure measures. Conducted at the Alzheimer Research Center, VU Medical Center, Amsterdam, the second study also enrolled 16 people with the same diagnosis who had evidence of brain amyloid deposition as per 11C PiB PET. This study administered either 40 mg or 125 mg VX-745 twice daily for 12 weeks, and measured the effect on safety and on brain amyloid burden, using a dynamic DVR protocol developed to reduce measurement variability in amyloid PET.

Results of both were presented at the 2016 CTAD and the 2017 AAIC meetings (Dec 2016 conference newsAug 2017 conference news). Neflamapimod caused no serious adverse events, and was reported to have shown signals on both amyloid removal and cognition, though interpreting open-label results of small trials versus practice effects remains questionable. The Amsterdam study was formally published (Scheltens et al., 2018).

December 2017 saw the start of REVERSE-SD, a Phase 2 study of 161 people with CSF biomarker-confirmed mild AD. This proof-of-concept trial compared a six-month course of a 40 mg neflamapimod capsule taken twice daily to placebo on change in total and delayed recall on the Hopkins Verbal Learning Test, Revised (HVLT-R); secondary outcomes include the Wechsler Memory Scale (WMS) Immediate and Delayed Recall, CDR-Sum of Boxes, MMSE, as well as CSF p-tau181 and Aβ1-40 (Alam et al., 2017). The trial took place at 38 study locations in the U.S., Europe, and the U.K.

On November 7, 2019, EIP Pharma announced that REVERSE-SD had failed to meet its primary endpoint of improving episodic memory. Neflamapimod was safe but performed like placebo on the HVLT-R, WMS, or any of the other secondary endpoints. A prespecified subgroup analysis found trends toward improvement in the HVLT-R and WMS in patients with the highest plasma drug concentrations. Compared with placebo, the treatment group had statistically significant reductions in the biomarkers CSF phospho-tau, total tau, and a trend toward reduced neurogranin (see press release). This data was subsequently presented at CTAD 2019 (Dec 2019 conference newsslides posted on company website), and published after peer review (Prins et al., 2021).

In October 2018, a fourth Phase 2 efficacy study began at University Hospital Toulouse. It enrolled 34 people with prodromal AD as per Dubois et al., 2014, whose memory impairment on the Free and Cued Selective Reminding test (FCSRT; Grober et al., 2008) falls into a defined range, and who have cerebral amyloidopathy as per CSF analysis or amyloid PET. This trial uses the microglial activation tracer DPA-714, which binds the microglial translocator protein TSPO, in an attempt to monitor brain inflammation in response to a 12-week course of treatment with 40 mg neflamapimod twice daily, or placebo. Three DPA-714 SUV measures represent the primary outcome; secondary outcomes span 35 measures ranging from neuropsychological assessments to blood and CSF markers of inflammation. The trial was completed in June 2021; no results have been made public.

In 2019, EIP Pharma began additional Phase 2 trials in people with dementia with Lewy bodies (DLB) and early Huntington's disease (HD). Called AscenD-LB, the first planned to recruit 80 people with DLB, who were taking cholinesterase inhibitors. The participants took 40 mg neflamapimod two or three times daily, based on weight, or placebo, for 16 weeks. The primary endpoint is change on a study-specific composite of six components of the Cogstate Neurological Test Battery, focused on attention, executive and visuospatial function. Secondary outcomes include the CDR-SB, MMSE, NPI, and International Shopping List Test, as well as a mobility test and electroencephalogram measures to evaluate potential biomarkers for DLB. The study was scheduled to take place at 22 sites in U.S. and two in the Netherlands, and run through mid-2020.

The second study was to enroll 16 people with genetically confirmed HD and cognitive deficits in Cambridge, England, U.K.; 15 were enrolled. Participants were to take 40 mg drug twice daily or placebo for 10 weeks, in a within-subject crossover design. They were evaluated on change in hippocampal function as assessed by a virtual human Morris Water Maze. Secondary outcomes included the CANTAB paired associates learning task and safety.

In November 2019, the FDA granted fast-track designation to neflamapimod for DLB.

On October 6, 2020, EIP Pharma announced that the AscendD-LB trial had met its primary endpoint (see company press release). According to results presented that November at CTAD, patients on neflamapimod three times daily improved on the NTB and the Attention Composite compared to those on twice-daily dosing or placebo. The gains were observed after four weeks of treatment, and maintained at 16 weeks. The same group improved on the secondary endpoint of the Timed Up and Go Test, which measures mobility and cognition. Other secondary endpoints of the NPI, CDR-SB, and International Shopping List Test trended toward improvement (Nov 2020 conference news). Results on the remaining secondary endpoints—MMSE and quantitative EEG—were not reported. The study enrolled 91 patients at 24 sites and had no withdrawals due to drug-related events, and no reported drug-related serious adverse events. According to final results presented at the November 2021 CTAD meeting, neflamapimod treatment resulted in slowing decline on the CDR-SB from baseline to week 16. A post hoc analysis found better drug effects in participants with low plasma ptau181, suggestive of “pure” DLB pathology, versus those with high ptau181, a sign of mixed pathology including amyloid.

Full trial data was published after peer review (Jiang et al., 2022). When the twice- and thrice-daily dosing groups were analyzed as a whole, their decline on the CDR-SB slowed compared to placebo. The treated group did better on the Timed Up and Go Test, a secondary endpoint. Cognitive measures of NTB, Attention Composite, and International Shopping List, which improved in the thrice-daily group, were no different from placebo when both dosing groups were combined. In this trial, only thrice-daily dosing achieved the targeted level of drug exposure. Twice-daily dosing resulted in lower blood levels than expected, and lower than estimated to be necessary for pharmacological activity. The analysis of ptau181 and drug response was published separately (Alam et al., 2023).

In October 2020, the HD trial was terminated before completion, due to COVID19 delays (see EU Clinical Trials RegisterEIP Pharma website). Results were posted on clinicaltrials.gov, and Huntington's was subsequently dropped as an indication from the company pipeline.

In May 2023, the company began another Phase 2 trial for DLB, comparing 16 weeks of thrice-daily 40 mg neflamapimod to placebo in 160 participants. Inclusion criteria differ slightly from the AscendD-LB study, in that participants must show loss of dopamine neurons on a PET scan, and have low CSF ptau-181. The CDR-SB is the sole primary outcome; secondary outcomes include the Timed Up and Go Test, NPI, and ADCS-Clinical Global Impression of Change. The study is running at 29 sites in the U.S. and the Netherlands, through August 2024.

For trials on this compound, see clinicaltrials.gov.

Clinical Trial Timeline

  • Phase 2
  • 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 2035
EIP Pharma, LLC NCT02423200
N=16
EIP Pharma, LLC NCT02423122
N=16

Last Updated: 06 Nov 2023

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Souvenaid

Tools

Back to the Top

Overview

Name: Souvenaid
Synonyms: Fortasyn Connect
Therapy Type: Supplement, Dietary (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Not Regulated)
Status in Select Countries: Marketed
Company: Nutricia

Background

Souvenaid is a marketed product. For sale in Australia and some European countries but not the U.S., Souvenaid is a medical food designed to enhance the formation and function of synapses (Oct 2009 conference newsde Wilde et al., 2011). It contains a combination of nutrient precursors and co-factors known to be needed in the synthesis of neuronal membranes. Called Fortasyn Connect, this formulation includes docosahexaenoic acid, eicosapentaenoic acid, uridine-5'-mono-phosphate, choline, phospholipids, selenium, and B, C, and E vitamins. 

Preclinical data on this formulation, or its components, were reported from more than a dozen studies in various cell-based and animal models. For example, components of Fortasyn Connect were reported to increase the brain concentration of synaptic proteins and phospholipids, and to stimulate neurite outgrowth and improve performance on behavioral assays in rats and gerbils (e.g. Wang et al., 2005Wurtman et al., 2006Cansev et al., 2008Holguin et al., 2008).

One study, by researchers at Nutricia, claimed a protective effect against Aβ42-induced toxicity in rats (de Wilde et al., 2011). Since then, work by affiliated researchers has reported that diets containing the components of Fortasyn Connect are neuroprotective, improve cerebral blood flow, and reverse spatial learning defects in the APP/PS1 mouse model of AD (Jansen et al., 2013; Zerbi et al., 2014; Koivisto et al., 2014).

Findings

Souvenaid is taken as a 125-ml breakfast drink. In clinical trials, it is being compared with a placebo of equal calories. 

In 2006, the first multicenter RCT, a 12-week study conducted in the Netherlands with 212 people with early AD was reported to have yielded an efficacy signal on one delayed verbal memory test but not on the ADAS-cog 13 (Aug 2008 conference news).

Starting in 2009, the multicenter S-Connect study evaluated a six-month course of Souvenaid in 527 people with mild to moderate AD who were on standard Alzheimer's therapy. The primary outcome measure was the ADAS-cog 11. Souvenaid was safe, and compliance was high as reflected by blood concentration of Souvenaid nutrient components; however, the treatment and placebo groups declined equally, showing no efficacy on the primary endpoint (Shah et al., 2013). 

Starting also in 2009, the Souvenir 1 and 2 trials respectively enrolled 225 and 255 patients with mild Alzheimer's disease who took no other AD medications. The former trial administered Souvenaid for three months and used the Wechsler Memory Scale test of delayed verbal recall, as well as the ADAS-cog 13, as its primary outcome; the latter trial ran for six months and used the z score on the neuropsychological test battery NTB as its primary outcome, with EEG measures of functional connectivity as a secondary outcome. The Souvenir 1 study reportedly showed a treatment benefit on a delayed verbal memory task, though the trial's other cognitive and functional outcome measures were unchanged (Scheltens et al., 2010). Souvenir 2 reported a trend toward benefit on the overall NTB, and a treatment benefit on the NTB's memory component.

Secondary analysis of 179 Souvenir 1 and 2 participants showed that during the treatment phase, β-band network measures of EEG differed between study groups in favor of the active group, indicating that Souvenaid might preserve synaptic brain networks, but the differences did not correlate with memory performance and their interpretation remains unclear. Souvenaid was again well-tolerated (Nov 2011 conference newsNov 2012 conference newsScheltens et al., 2012de Waal et al., 2014). In a six-month, open-label extension offered to patients in both trials, data from 201 participants confirmed high compliance and safety, as well as a treatment benefit on an exploratory NTB readout (Olde Rikkert et al., 2015). In a meta-analysis of Souvenir 1, 2, and S-Connect data, scientists concluded that the supplement achieved a clinically detectable effect in patients with early AD (Cummings et al., 2017).

In 2009, the LipiDiDiet Trial began enrolling 300 people with prodromal Alzheimer's who do not take other AD medications for a two-year trial, with up to four additional years of optional, blinded extensions. The primary outcome was the NTB, and secondary outcomes included progression to dementia, other cognitive and functional measures, blood markers, and brain atrophy. This trial ran at sites in Finland, Germany, the Netherlands, and Sweden. It was completed in early 2019.

After two years of use, Souvenaid did not affect performance on the NTB, but did lead to slower decline on the CDR-sb and less hippocampal shrinkage (Mar 2016 conference news; Soininen et al., 2017). A post hoc analysis indicated that people on Souvenaid slowed their cognitive decline by one-third when measured on the ADCOMS (Hendrix et al., 2019).

At the 2020 AAT-AD/PD meeting, investigators reported results of a one-year extension including 45 treated and 36 placebo participants. After three years, the treatment effect widened. Those on Souvenaid showed a 45 percent slowing of decline on the CDR-sb compared with placebo, and a 33 percent decline in hippocampal atrophy. Both were significant (Apr 2020 conference news). A subsequent publication of the results reported improvement on additional endpoints, including a 60 percent slowing of decline on the NTB five-item composite, and 76 percent slowing on the NTB memory domain, compared to placebo. Whole brain atrophy slowed by 22 percent (Soininen et al., 2020).

In 2012, an RCT began to evaluate the effects of four weeks of once-daily Souvenaid on brain metabolism by way of brain magnetic resonance spectroscopy. It enrolled 34 people with mild to moderate AD. Compared with placebo controls, people on Souvenaid had changes in indices of phospholipid synthesis and breakdown, and choline metabolism; markers of neural integrity and gliosis were unchanged (Rijpma et al., 2017).

In 2013, the open-label AWARE study began to enroll 315 people with AD to evaluate the effect of Souvenaid in clinical practice settings against the Amsterdam version of the International Activities of Daily Living Questionnaire. According to a poster presented at the 2020 AAIC conference, the study enrolled 116 people, who declined less on the IADL after six or 12 months of Souvenaid than a reference population (Ziere et al., 2020). 

In 2014, NL-ENIGMA began comparing six months of Souvenaid with placebo on the outcome of glucose metabolism as assessed by FDG-PET of brain regions affected in Alzheimer's disease (Scheltens et al., 2016). The trial enrolled 50 people with prodromal or mild AD; it found no change within treatment groups and no difference between treatment groups in glucose metabolism (Scheltens et al., 2019). A separate study reported that one year of Souvenaid significantly preserved glucose uptake on FDG-PET in people with MCI (Palomo et al., 2019).

Souvenaid has also been reported to reduce behavioral symptoms in a small, three-month crossover trial of frontotemporal dementia (Pardini et al., 2015).

In November 2019, a Phase 2 trial was registered that will compare one year of daily Souvenaid with a vitamin-only placebo on cognitive decline during normal aging. To be conducted at the University of Miami, this trial was originally scheduled to begin in December 2019 to enroll 120 participants aged 55 and 89 with age-related cognitive decline determined by the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), and a screen for subjective cognitive decline. Primary measures include a variety of cognitive tests. The study is now slated to begin in February 2021.

A trial will begin in April 2021 to study the feasibility of Souvenaid supplementation after acute traumatic brain injury. The primary outcome of the 50-person study will be retention rate after six months.

Souvenaid will be used in MIND-AD, a large, multimodal prevention trial planned for people with prodromal AD in Sweden, Finland, France, and Germany (see website). A pilot trial began in October 2017 to recruit 150 participants between 60 and 85 years old with biomarker-confirmed AD, low normal scores on memory tests, and potential for lifestyle improvements. Participants will follow one of three programs: regular health advice, a multidomain lifestyle intervention including nutritional guidance, exercise, cognitive training and management of vascular and metabolic risk factors, or the lifestyle intervention plus Souvenaid. The trial will run six months, with an optional six-month extension. The primary outcome is feasibility, assessed by recruitment time, overall adherence, and retention rate. The trial was set to end in November 2020.

In December 2020, a Cochrane meta-analyses concluded that Souvenaid "probably does not reduce the risk of progression to dementia in people with prodromal AD." Also, that "There is no convincing evidence that Souvenaid affects other outcomes important to people with AD in the prodromal stage or mild to moderate stages of dementia. Conflicting evidence on combined cognitive-functional outcomes in prodromal AD and mild AD dementia warrants further investigation" (Burckhardt et al., 2020).

For a listing of trials, see International Clinical Trials Registry Platform. For details on recent trials, see clinicaltrials.gov.

Last Updated: 17 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

BI 409306

Tools

Back to the Top

Overview

Name: BI 409306
Synonyms: SUB 166499
Therapy Type: Small Molecule (timeline)
Target Type: Other Neurotransmitters (timeline), Other (timeline)
Condition(s): Alzheimer's Disease, Schizophrenia
U.S. FDA Status: Alzheimer's Disease (Discontinued), Schizophrenia (Phase 2)
Company: Boehringer Ingelheim

Background

BI 409306 is an inhibitor of phosphodiesterase 9A. PDE9A is a drug target for cognitive decline in Alzheimer's and other diseases because this enzyme reduces brain levels of the second messenger cyclic guanosine monophosphate. cGMP transduces signals by the neurotransmitters nitric oxide and glutamate. This pathway modulates synaptic transmission and plasticity in the hippocampus and cerebral cortex, and is reduced in AD brain (Zhihui 2013).

No peer-reviewed publications on BI 409396 preclinical data are published, but Boehringer Ingelheim published at the 2015 AAIC conference that BI 409306 increased hippocampal LTP and improved performance in an episodic memory task in rats (Rosenbrock et al., 2015).  Prior, and since terminated, PDE9A inhibitors have been reported to increase CSF levels of cGMP and to increase hippocampal expression of the glutamate receptor subunit GluR1. They were reported to affect indicators of synaptic plasticity, such as increasing LTP in rat hippocampal slices and enhancing memory and attention on a range of behavioral assays in rats (van der Staay et al., 2008Hutson et al., 2011Kroker et al., 2012Vardigan et al., 2011). Research on therapeutic PDE9A inhibitors goes back to at least 2005 (Wunder et al., 2005).

Findings

As of spring 2018, Boehringer Ingelheim had conducted 16 Phase 1 studies of this compound, testing both tablets and liquid formulations in a total of about 550 people in Germany, Belgium, South Korea, and the United States. Fourteen studies were in healthy volunteers, two in people with Alzheimer's disease or schizophrenia. The studies assessed safety, tolerability, pharmacokinetics and -dynamics of single or ascending doses given once or twice daily, as well as drug-drug interactions and the effect of food on BI 409306's metabolism. Company researchers reported at AAIC that the compound did not interact with donepezil (Wunderlich et al., 2017).

Three recent Phase 1 trials, initiated in 2015 and 2017, evaluate cardiac effects, disorders of the eye, and drug interactions with the antibiotic rifampicin, respectively. At AAIC 2015, Boehringer Ingelheim presented data from a prior Phase 1 trial in Japanese and Chinese healthy male volunteers, which noted light phobia and other visual adverse effects (Wunderlich et al., 2015, available as AAIC abstract on company website). Safety and pharmacokinetic data on 80 healthy volunteers were formally published (Moschetti et al., 2016).

In December 2014 and January 2015, respectively, two three-month, multinational Phase 2 trials of BI 409306 began in patients with prodromal AD. One study compared four doses of BI 409306 to placebo in 288 people with mild to moderate Alzheimer's who have not taken a cholesterase inhibitor or memantine in the past three months; the other compared the same doses as add-on therapy in 336 patients who also take donepezil. For both studies, the primary outcome measure was the neuropsychological test battery (NTB); secondary outcomes included the ADCS-MCI-ADL, CDR-SB, ADAS-Cog11, and ADCS-ADL (Wunderlich et al., 2016). In February 2018, Boehringer Ingelheim announced topline results from both trials, for which data were pooled. There was no difference between drug and placebo groups on the primary outcome, and the company decided to stop developing this compound for AD (see Feb 2018 news).

BI 409306 continues being evaluated in schizophrenia. Results of one Phase 2 trial, which missed its primary endpoint of cognitive improvement, are published (Sand et al., 2017).

For all trials of this compounds, see clinicaltrials.gov.

 

Clinical Trial Timeline

  • Phase 2
  • 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 2035
Boehringer Ingelheim NCT02240693
N=128
Boehringer Ingelheim NCT02337907
N=329

Last Updated: 25 May 2018

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

S 38093

Tools

Back to the Top

Overview

Name: S 38093
Therapy Type: Small Molecule (timeline)
Target Type: Cholinergic System (timeline), Other Neurotransmitters (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Discontinued)
Company: Servier

Background

This histamine H3 receptor antagonist was being developed for the treatment of Alzheimer's disease in many European, South American, and other countries, though not the United States. Blocking the histamine H3 receptor has been a target of several small-molecule therapeutic programs for cognition disorders at other companies, as well, e.g. GSK239512ABT-288. The approach has been proposed to increase cholinergic signaling, which wanes with neurodegeneration in AD (Passani and Blandina, 1998Bembenek et al., 2008Bertoni et al, 2008). 

No peer-reviewed studies are published on this compound; however, an AAIC meeting presentation on S 38093 reported benefits on attention and executive function in five monkeys treated with the dopaminergic toxin MPTP (Schneider et al, 2009).

Findings

Between 2009 and 2015, four Phase 2 trials, testing doses ranging from 2 to 25 mg of S 38093 per day have been conducted in a total of about 1,600 patients with mild to moderate Alzheimer's disease. Two studies compared S 38093 monotherapy tablets to placebo, and two compared the same doses of S 38093 as an add-on to standard cholinesterase inhibitor therapy.

The most recent trial started in 2012. It was an international Phase 2b study evaluating 2, 5, or 20 mg per day of S 38093 co-administered with 10 mg of donepezil, given for six months, to donepezil alone in 700 patients with moderate AD. Outcomes included a cognitive benefit as measured by the ADAS-cog 11 and a functional benefit as measured by DAD. No biomarkers were used in this study. This trial was completed in January 2015. 

At the November 2015 CTAC conference, S 38093 development was reported to have been discontinued due lack of efficacy in Phase 2. Neither of the three doses, taken for a year, outperformed placebo on any measure of cognition, function, clinical global impression, or caregiver burden (see Nov 2015 conference news).

For all Phase 2 trials of this compound, see EU Clinical Trials Register.

Last Updated: 15 Jan 2016

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Umibecestat

Tools

Back to the Top

Overview

Name: Umibecestat
Synonyms: CNP520, 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: Amgen, Inc., Novartis Pharmaceuticals Corporation

Background

CNP520 is an oral, small-molecule inhibitor of the aspartyl protease BACE designed to reduce Aβ production to prevent or treat Alzheimer's disease. BACE1 is the β-secretase enzyme that cleaves the APP protein to release its C99 fragment, which gives rise to various species of Aβ peptide during its cleavage by γ-secretase. The rationale of BACE inhibition is that it represents an upstream interference with the amyloid cascade, regardless of which species or aggregation states of Aβ then exert toxicity in the brain. BACE inhibition is sometimes envisioned as long-term maintenance therapy to limit Aβ production after an initial round of immunotherapy to remove existing amyloid deposits. 

Developed by Novartis, CNP520 is being jointly developed with Amgen (see company press release). CNP520 is formulated to be taken as capsules.

At the 2016 AAIC conference in Toronto, preclinical data presentations reported significant selectivity of CNP520 for BACE1 over BACE2 and other proteases such as renin and cathepsin, as well as absence of skin discoloration seen with some prior BACE inhibitors (see Aug 2016 conference news).

Findings

No Phase 1 trials for this compound are listed in clinicaltrials.gov; however, at the 2016 AAIC conference, Novartis scientists presented safety, tolerability, and pharmacodynamic data from three Phase 1 trials in a total of 113 healthy volunteers. The presentation also included data on dose-dependent lowering of CSF Aβ40 by up to 95 percent (see Aug 2016 conference news).

From August 2015 to March 2016, Novartis ran a multisite Phase 2a dose-ranging trial in the Netherlands, Belgium, Germany, the United Kingdom, and the United States. It evaluated safety and tolerability, as well as plasma and CSF pharmacokinetics and pharmacodynamics of 2 mg, 10 mg, 35 mg, and 85 mg of CNP520 or placebo taken once daily for 13 weeks by 124 healthy men and women between 60 and 80 years of age. The trial measured change in CSF Aβ38, 40, and 42. Results are posted on clinicaltrials.gov. Structural features of CNP52, extensive preclinical data in rodents and dogs, as well as data from four Phase 1 trials and this Phase 2a trial were published in a peer-reviewed journal (Neumann et al., 2018).

In November 2015, a secondary prevention study called GENERATION 1 started enrolling 1,340 cognitively normal, homozygous ApoE4 carriers age 60 to 75. Part of the Alzheimer's Prevention Initiative (API) in partnership with Banner Alzheimer's Institute, this Phase 2/3 study is set to run until 2024 (Lopez Lopez et al., 2017Lopez Lopez et al., 2019). The treatment period is five years. This study will randomize half of participants to compare once-daily 50 mg CNP520 to matching placebo, the other half to compare quarterly injections of the investigational active immunotherapy CAD106 to placebo. The trial will measure the agents' ability to delay diagnosis to MCI or AD dementia and change on the APICC cognitive composite (Langbaum et al., 2015). An extensive list of secondary outcomes includes change on the Clinical Dementia Rating Scale sum of boxes (CDR-SB) and other cognitive/functional scales, fluid biomarkers including CSF Aβ and tau, brain imaging including volumetric MRI plus amyloid PET and tau PET measurement of brain amyloid and tangle deposition, respectively, as well as safety measures and Aβ titers (see Jul 2014 conference news).

In August 2017, Novartis and API/Banner Alzheimer’s Institute began an additional Phase 2/3 prevention study. GENERATION 2 is aiming to enroll 2,000 cognitively normal homozygous ApoE4 carriers, or heterozygous carriers with evidence of brain amyloid, age 65 to 70. Participants are to be randomized to one capsule of 15 or 50 mg CNP520 or placebo daily, for at least 60 months, and up to a maximum of 84 months. Like in GENERATION 1, the primary outcome is time to a diagnosis of MCI or dementia due to AD or change on the APICC cognitive composite. Ten secondary outcomes include change on the CDR-SB, RBANS, and other cognitive/functional scales, fluid biomarkers including CSF Aβ and tau, brain imaging including volumetric MRI and measures of cerebral amyloid angiopathy, plus amyloid PET and tau PET measurement of brain amyloid and tangle deposition, respectively, as well as safety measures. This trial is set to run until 2025.

GENERATON trial participation requires disclosure of ApoE genotype to participants and, in the case of GENERATION 2, of brain amyloid status, as well.

On July 11, 2019, the trial sponsors announced a premature end to CNP520 dosing in both GENERATION trials, citing cognitive worsening in the treatment groups measured during a preplanned interim analysis (July 2019 conference news). Participants taking CNP520 subtly declined on the RBANS cognitive composite, had more brain atrophy and lost more weight than people on placebo (Dec 2019 conference news). The results were similar to those of other BACE inhibitors, verubecestat and atabecestat). Researchers continued to monitor cognition and brain volume after treatment was stopped, and reported that cognitive deficits and brain volume loss were reversible (Aug 2020 conference news).

For all trials of this compound, see clinicaltrials.gov and EU Clinical Trials Register.

Clinical Trial Timeline

  • Phase 2
  • Phase 2/3
  • 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 2035
Novartis Pharmaceuticals Corporation NCT02576639
N=125
Novartis Pharmaceuticals Corporation NCT02565511
N=1340
Novartis Pharmaceuticals Corporation NCT03131453
N=2000

Last Updated: 21 Jan 2021

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

rTgTauEC

Synonyms: neuropsin-tTA x FVB-Tg(tetO-tauP301L)4510

Tools

Back to the Top

Species: Mouse
Genes: MAPT
Modification: MAPT: Transgenic
Disease Relevance: Frontotemporal Dementia, Alzheimer's Disease
Strain Name: N/A

Summary

Phenotype Characterization

When visualized, these models will distributed over a 18 month timeline demarcated at the following intervals: 1mo, 3mo, 6mo, 9mo, 12mo, 15mo, 18mo+.

Absent

  • Plaques

No Data

Plaques

Absent.

Tangles

By 18 months of age, Gallyas silver-positive staining is observed, indicative of paired helical filaments. This is followed by thioflavin-S staining at 24 months. Tau pathology develops first in neurons of the medial EC expressing human tau, followed by neurons in the dentate gyrus, CA1 and CA2/3(de Calignon et al., 2012).

Synaptic Loss

By 24 months of age pre- and post-synaptic densities were reduced in the middle third of the molecular layer of the dentate gyrus as measured by synapsin-1 and PSD-95 staining (de Calignon et al., 2012).

Neuronal Loss

Neuronal loss is detectable by 24 months of age in areas with transgene expression (e.g. layer II of the EC and parasubiculum), compared with age-matched mice expressing only tTA. Significant neuronal loss was not observed at 21 months (de Calignon et al., 2012).

Gliosis

Microglial activation and astrogliosis by 24 months of age, in conjunction with axonal degeneration and neuronal loss (de Calignon et al., 2012).

Changes in LTP/LTD

At 16 months of age, subtle differences in electrophysiological properties have been observed in the perforant pathway, including a decrease in LTP and an increase in the probability of neurotransmitter release (Polydoro et al., 2014).

Cognitive Impairment

Very mild and specific deficits in contextual fear conditioning at 16 months of age, but no deficits in the radial arm maze (Polydoro et al., 2014).

Last Updated: 13 Apr 2018

COMMENTS / QUESTIONS

  1. Well planned and well done!

    View all comments by Takaomi Saido

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Subscribe to ALZFORUM RSS