Therapeutics

Low Dose Interleukin-2

Tools

Back to the Top

Overview

Name: Low Dose Interleukin-2
Synonyms: COYA-301, Aldesleukin, Proleukin, COYA-302, ld IL-2
Therapy Type: Other
Target Type: Inflammation (timeline)
Condition(s): Alzheimer's Disease, Amyotrophic Lateral Sclerosis
U.S. FDA Status: Alzheimer's Disease (Phase 2), Amyotrophic Lateral Sclerosis (Phase 3)
Company: Coya Therapeutics

Background

Low-dose interleukin 2 is being tested as an immunomodulatory approach in neurodegenerative diseases. It increases the number and activity of regulatory T cells (Tregs) that suppress inflammation. At high doses, IL-2 stimulates the immune response to tumors, and is approved for treating cancer.

Treg-mediated immune control appears to be compromised in people with AD (Faridar et al., 2020). In preclinical studies, Tregs that were transferred into AD mice entered the brain and stopped Aβ plaque growth (Faridar et al., 2022). Multiple labs have reported that low-dose IL-2 induces expansion of Treg cells, decreases hippocampal Aβ plaque load, and lessens memory deficits in APP/PS1 mice (Dansokho et al., 2016; Alves et al., 2017; Yuan et al., 2023).

Tregs also play a role in ALS and in Parkinson’s disease. In people with ALS, an observed decline in Tregs correlated with faster disease progression. In the SOD1 mouse model of ALS, giving IL-2 or Tregs resulted in motor neuron preservation, less spinal cord inflammation, and longer survival (Sheean et al., 2018; Banerjee et al., 2008). A Phase 1 study in ALS patients demonstrated reduction in markers of oxidative stress and inflammation after Treg infusion (Beers et al., 2022).

In PD patients, reductions in Treg number and function correlated with proinflammatory T cell activation, and increases in Tregs were associated with improved clinical measures (Thome et al., 2021; Arce-Sillas et al., 2024). Administration of low-dose IL-2 led to Treg expansion in a mouse model of PD (Markovic et al., 2022). In other preclinical studies, transplantation or expansion of Tregs was neuroprotective in rodent models of PD (Reynolds et al., 2010; Badr et al., 2022; Park et al., 2023).

Findings

In June 2019, academic investigators in Houston, Texas, began an open-label feasibility study of low-dose commercially available recombinant human IL-2 in people with AD dementia. Eight participants injected 1 million units of Aldesleukin daily for five days per month, for four months. Results are published (Faridar et al., 2023). The treatment was safe and well-tolerated. Most common adverse events were injection site reactions and mild leukopenia, each affecting one-third of patients. After each cycle, participants had increased Treg numbers and function, which returned to baseline between treatments. IL-2 treatment downregulated inflammatory cytokine expression in circulating monocytes, and some plasma pro-inflammatory markers decreased compared to baseline. The investigators claimed an improvement on the MMSE that reverted to baseline after treatment had ended. ADAS-Cog and CDR-SB scores did not change.

In 2020, COYA Therapeutics was founded by the Houston researchers to develop low-dose IL-2 under the name COYA-301. The company presented more results of the open-label AD study in May 2023, indicating a significant lowering of monocyte inflammatory mediators TNFα, IL-6, and IL-1β after treatment. In one patient, TSPO-PET scans suggested a reduction in brain inflammation two weeks after the last dose (slides).

In January 2022, Phase 2 began with support from the Gates Foundation and the Alzheimer’s Association. The Houston investigators enrolled 38 AD patients with mild to moderate dementia, for six months of Aldesleukin, given as a five-day course by subcutaneous injection once or twice per month. The primary endpoint was safety and tolerability. Change in Tregs as a percentage of total CD4 T cells served as a secondary endpoint. In May 2024, COYA announced completion of the study, with results to follow later in the year (press release).

In September 2022, an independent Phase 2 trial began enrolling 45 AD patients with mild dementia at the Centre Hospitalier St. Anne in Paris. Treatment consists of 1 million units of IL-2 or placebo, on a schedule of daily injections for five days, followed weekly injections for four months. The primary endpoint is change in Clinical Dementia Rating 18 months after the first injection. Other endpoints include the MMSE, ADAS-Cog, ADCS-ADL, CDR-SB, change in Tregs and other immune cells, PET scans for neuroinflammation, hippocampal atrophy, and safety. The trial will run until September 2025.

In March 2023, a biomarker study began to assess the effects of low-dose IL-2 on CSF and blood markers of inflammation, and on brain inflammation using the ER176 PET tracer that binds TSPO. This placebo-controlled study will enroll 40 patients in Houston, to receive IL-2 every two or four weeks for six months. The study will run until December 2025.

Testing of low-dose IL-2 for ALS began in 2015, with a Phase 2 trial by researchers in Nimes, France. Thirty-six patients received 1 or 2 million units of IL-2, or placebo, by subcutaneous injection for five days each month for three months, in addition to riluzole. IL-2 was tolerated, with mild adverse events that included injection site reactions and flu-like symptoms. The study achieved its primary outcome of increasing Tregs in the treatment groups. The plasma inflammation marker CCL2 decreased dose-dependently, but there was no change in the surrogate efficacy marker of plasma neurofilament light (Camu et al., 2020). Analysis of leucocyte gene expression showed a dose-dependent increase in Treg markers at the end of treatment (Giovanelli et al., 2021). Inhibition of inflammatory gene expression was apparent after the first cycle of treatment, and was less pronounced after three cycles. Higher baseline inflammatory gene expression predicted poorer response to treatment.

From 2017-2021, researchers followed up with a larger Phase 2 trial in France and the U.K. The MIROCALS study enrolled 220 patients newly diagnosed with ALS, for five-day courses of 2 million units Il-2 per day, or placebo, monthly for 18 months. The primary outcome was survival. Results were announced in December 2022 (press release). Treatment was safe, tolerated, and resulted in elevated Treg numbers. Treatment was associated with a non-significant 19 percent reduction in the risk of death. In a prespecified subgroup analysis, participants with low CSF phosphorylated neurofilament heavy chain and less aggressive disease had a significant 40 percent increase in survival. This subgroup included most of the participants in the trial.

From 2019-2022, investigators in Houston and Boston tested the combination of monthly injections of patient-derived Tregs with low dose IL-2 in 12 ALS patients. The treatment was safe and resulted in elevations of Treg suppressive function (Thonhoff et al., 2022).

In 2020, another open-label trial began recruiting 13 ALS patients in China; its status is unknown.

In October 2021, COYA started an open label study in people with ALS of low-dose IL-2 in combination with CTLA4-IgG. Called Abatacept, this drug suppresses the activation of monocytes and microglia; it is used to treat autoimmune diseases. The company is testing this dual immunomodulator under the name COYA-302. The study, at the Houston Methodist Research Institute, planned to treat 10 patients every two weeks for one year, and assess safety and tolerability. Secondary and exploratory outcomes include Treg function, serum biomarkers of oxidative stress, inflammation, and neurodegeneration, and clinical functioning on the ALSFRS-R scale. Results are published (Thonhoff et al., 2024). Four patients enrolled; all completed it without serious adverse events. Increases in Treg in number and function occurred over the entire treatment period, and returned to baseline levels after treatment stopped. Biomarkers showed trends to reduction in the first 16 weeks, but were not uniformly changed. ALSFRS-R scores remained stable over 48 weeks.

In May 2024, the Houston researchers began a small trial testing COYA-302, dosed every two or four weeks, in 10 frontotemporal dementia patients. The open-label treatment will run for six months with a primary outcome of safety, and a secondary outcome of blood Treg numbers. Completion is expected in April 2026.

For details on low dose IL-2 trials, see clinicaltrials.gov.

Last Updated: 28 Jun 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

Vps35 p.D620N KI Mouse

Tools

Back to the Top

Species: Mouse
Genes: Vps35
Modification: Vps35: Knock-In
Disease Relevance: Parkinson's Disease
Strain Name: B6.Cg-Vps35tm1.1Mjff/J

Modification Details:

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

  • Non-Motor Impairment

No Data

Neuronal Loss

Loss of TH-positive neurons in the SNpc and loss of TH-positive nerve terminals in the striatum at 13-16 months. Widespread axonal degeneration in the brain at 13 months.

Dopamine Deficiency

Enhanced peak amplitude in dopamine release and prolonged reuptake kinetics in acute striatal slices from 3-month-old mice; decreased DAT and increased VMAT2. Basal levels of dopamine and metabolites in dorsolateral striatum did not differ, but the DOPAC+HVA/DA ratio was increased. At 16 months, dopamine in striatal homogenates was reduced, but levels of metabolites (DOPAC, HVA) did not differ.

α-synuclein Inclusions

No differences in α-synuclein puncta density or distribution in the SNpc at 3 months. No pathological α-synuclein observations seen throughout the brain at 13 months. However, at 15 to 16 months, increased somatic α-synuclein immunoreactivity found in the SNpc, and increased α-synuclein oligomers and aggregated α-synuclein observed in the ventral midbrain.

Neuroinflammation

Increased GFAP immunostaining in the SNpc, but not in the striatum, of 15- to 16-month-old VKI mice; no GFAP differences observed at earlier ages. No differences in microgliosis (Iba-1 immunostaining) in the SNpc or the striatum up to 16 months of age.

Mitochondrial Abnormalities

Mitochondrial structure, assesed by EM, was perturbed at 14 months, but not at 3 months, of age. Mitochondrial function—namely, the oxygen consumption rate—was reduced in older (15-month-old) mice.

Motor Impairment

Motor deficiencies on the open-field test and the beam walking test appear at 14 months of age, but not earlier from 3 to even 13 months of age. However, performance on other motor tests—Rotarod and grip strength—did not differ at the advanced age (14 months). No deficits seen in the cylinder test (rearing) at 3 months. Amphetamine-induced hyperlocomotion is rescued by LRRK2 kinase inhibition.

Non-Motor Impairment

No deficits in the buried pellet test, measuring olfactory function, from 6 to 14 months of age. No defects in gastrointestinal function (as measured by stool frequency and water content) up to 14 months of age.

Last Updated: 17 Jun 2024

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

Gba1 D409V KI Mouse (MJFF)

Synonyms: Gba D409V KI   , Gbatm2636(D427V)Arte

Tools

Back to the Top

Species: Mouse
Genes: Gba1
Modification: Gba1: Knock-In
Disease Relevance: Parkinson's Disease
Strain Name: C57BL/6N-Gba1tm1.1Mjff/J

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

  • Neuronal Loss

No Data

  • Mitochondrial Abnormalities

Neuronal Loss

No differences in the number of dopaminergic neurons in the substantia nigra pars compacta were found between homozygous KI mice and wild-type mice at 4, 8, and 12 months of age.

Dopamine Deficiency

Dopamine levels did not differ at 4, 8, and 12 months of age, but dopamine turnover (ratio of DOPAC and HVA to dopamine) tended to increase, though the increase was only significant at 12 months of age.

α-synuclein Inclusions

Homozygous KI mice have higher levels of soluble monomeric α-synuclein in the hippocampus at 12 months than heterozygous KI mice and wild-type controls. Levels of pathologic phosphorylated form pS129 do not differ between homozygous KI mice and controls in the substantia nigra, cortex, or hippocampus.

Neuroinflammation

Data are mixed on levels of GFAP and Iba-1 immunostaining in KI mice brain. One study in homozygous KI mice found no differences in the striatum and substantia nigra at 4, 8, or 12 months of age; another found decreased GFAP staining in the substantia nigra at 12 months; and a third study (het mice) found increased GFAP and Iba-1 in the hippocampus at 12 months.

Mitochondrial Abnormalities

No data.

Motor Impairment

Homozygous D409V KI mice generally exhibit motor function similar to wild-type controls (open-field, Rotarod, grip strength, swim velocity). However, a couple of exceptions found in one study were greater grip strength force at 12 months of age and transiently increased locomotor activity on the open-field test at 8 months of age.

Non-Motor Impairment

Cognitive performance was impaired in 12-month-old heterozygous KI mice (but not at 3, 6, or 9 months), based on the Morris water maze and Y-maze. Anxiety-like behavior (based on the open-field test) did not differ at 12 months.

Last Updated: 14 Jun 2024

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

VY-TAU01

Tools

Back to the Top

Overview

Name: VY-TAU01
Synonyms: HC3LC2, Voyager_9
Therapy Type: Immunotherapy (passive) (timeline)
Target Type: Tau (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 1)
Company: Voyager Therapeutics

Background

VY-TAU01 is a recombinant, humanized IgG4 monoclonal antibody to an epitope in the C-terminus of tau. It is designed to block the spread of pathological tau.

According to a poster presented at AAIC 2022, this antibody was generated by immunizing mice with paired helical filamentous tau isolated from human brain. More than 700 monoclonal antibodies were screened for selectivity to pathological tau, as well as for their ability to inhibit seeding of tau aggregates in vitro and spreading of tau pathology in mice. Voyager presented data demonstrating that the most efficacious C-terminal antibody, Ab01, inhibited the spread of pathological tau by greater than 70 percent in the P301S mouse seeding model. This mouse IgG1 antibody was subsequently humanized to generate the clinical candidate (AD/PD 2023 conference poster).

In nonhuman primates, blood levels after intravenous administration were dose-proportional. The antibody’s half-life was 11-12 days, CSF exposure was approximately 0.1-0.2 percent of serum (AD/PD 2024 poster). There was no indication of a significant anti-drug antibody response.

Findings

On May 16, 2024, Voyager announced the start of clinical development with a single-ascending-dose safety and pharmacokinetic study of intravenous VY-TAU01 (press release). The trial expects to enroll 48 healthy volunteers in multiple dose cohorts. The trial was not found in registries.

Last Updated: 03 Jun 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

NX210c

Tools

Back to the Top

Overview

Name: NX210c
Synonyms: HWSGWSS[CSRSC]GOH (brackets represent the disulfide bond)
Chemical Name: H-L-tryphophanyl-L-seryl-glycyl-L-tryptophanyl-L-seryl-L-seryl-L-cysteinyl-L-seryl-L-arginyl-L-seryl-L-cysteinyl-glycyl-OH (disulfide bond)
Therapy Type: Small Molecule (timeline)
Target Type: Other (timeline)
Condition(s): Amyotrophic Lateral Sclerosis, Parkinson's Disease
U.S. FDA Status: Amyotrophic Lateral Sclerosis (Phase 2), Parkinson's Disease (Phase 1)
Company: Axoltis Pharma

Background

NX210c is a cyclized 12-amino-acid peptide derived from a protein involved in embryonic neuronal development. It is delivered by intravenous injection.

This peptide was designed from the most conserved sequence of the thrombospondin repeats from the SCO-spondin protein. SCO-spondin, a large, multifunctional glycoprotein specific to the CNS extracellular matrix, is produced by the subcommissural organ during embryogenesis, and is essential for neural development. SCO-spondin has also been proposed to function in adult neurogenesis (Inada et al., 2023).

Both linear and cyclic forms of this peptide have been evaluated preclinically. The linear NX210 is converted in vivo to NX210c by formation of an intrachain disulfide bridge between two cysteines. In a mouse amyloidosis model, NX210 and NX210c reduced pathologic markers of Aβ42, p-Tau, and inflammation, after intraventricular injection of Aβ oligomers (Le Douce et al., 2021). In this study, conducted by Axoltis researchers, the peptides improved behaviors related to cognition and memory in the Y maze, passive avoidance test, and Morris water maze.

In other Axoltis-sponsored preclinical work, NX210 protected against oxidative stress and stimulated axonal regrowth and functional recovery in a spinal cord injury model (Sakka et al., 2014). NX201 and NX201c both protected neurons from glutamate excitotoxicity by disrupting the apoptotic signaling cascade; NX210c was more effective (Deletage et al., 2021). NX210c increased synaptic transmission in brain slices, and improved memory in mice treated with the NMDAR agonist phencyclidine (Lemarchant et al., 2022). The company also claims that NX210c reduces blood-brain barrier permeability, but that data is not public. In the only published study independent of Axoltis, NX210 protected against cell loss in ischemic stroke models (Yang et al., 2022).

Findings

From June to November 2020, the company conducted a Phase 1 safety trial of NX210. It enrolled 39 healthy adults in five cohorts, to receive single intravenous doses of 0.4, 1.25, 2.5, 5, or 10 mg/kg, or placebo. According to published results, one-third of participants reported mild adverse events. The most common were dizziness, headache, and sleepiness. NX210 cyclized to NX210c in blood. The peptide was widely distributed and rapidly cleared, with a half-life in plasma of 20 minutes or less. Potential pharmacodynamic effects were reported on EEG, and on plasma tryptophan and homocysteine (Bourdes et al., 2022).

In October 2022, the FDA granted NX210c Orphan Drug Designation for ALS.

In December 2022, a multiple-dose Phase 1 trial began enrolling 29 healthy aged volunteers. Thirty participants are to receive 5 or 10 mg/kg NX210c or placebo by 10-minute intravenous infusion three times a week for four weeks. The primary outcome is adverse events; blood and CSF pharmacokinetic measures serve as secondary outcomes. The study assesses neurologic safety using the Neurocart Battery, as well as 34 blood and 16 CSF biomarkers. Conducted in the Netherlands, the trial is set to finish in December 2024.

In December 2023, the company announced initial results (press release). The treatment remained safe and well-tolerated, with mild adverse events and no evidence of neurologic harm. Data presented at the 2024 AD/PD conference in Lisbon claimed decreases in blood homocysteine and the tight junction protein claudin 5, which were interpreted as evidence of blood-brain barrier repair. Blood levels of neurofilament light chain were said to decrease, although no numbers were shown. Changes in blood and CNS biomarkers were sustained for 40 days of follow-up, despite the drug’s short plasma half-life. The company plans to expand this trial to enroll people with Parkinson’s disease.

In April 2024, the company registered a Phase 2 trial  in ALS. To begin in September 2024, it will recruit 80 adult patients. The trial will compare four weeks of thrice-weekly 5 or 10 mg/kg doses to placebo, with a three-month follow-up. The sole outcome listed on clinicaltrials.gov is blood neurofilament light, or the CSF/serum albumin ratio, an indicator of blood-brain barrier integrity. Completion is expected in February 2026.

For details on NX210c trials, see clinicaltrials.gov and the WHO Trial Registry.

Last Updated: 03 Jun 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Selnoflast

Tools

Back to the Top

Overview

Name: Selnoflast
Synonyms: RO7486967, RG-6418, IZD-334, somalix
Chemical Name: 1-ethyl-N-[(1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl]piperidine-4-sulfonamide
Therapy Type: Small Molecule (timeline)
Target Type: Inflammation (timeline)
Condition(s): Parkinson's Disease
U.S. FDA Status: Parkinson's Disease (Phase 1)
Company: Hoffmann-La Roche, Inflazome Ltd.

Background

Selnoflast is an inhibitor of inflammasomes containing NLRP3, or nod-like receptor family, pyrin domain-containing protein 3. Inflammasomes are multiprotein, cytosolic complexes that function as sensors in the innate immune system. Their activation by pathologic proteins and other stressors triggers production and secretion of proinflammatory cytokines IL1-β and IL-18, and can induce cell death. NLRP3-containing inflammasome activation occurs in many conditions where chronic inflammation plays a role, including Alzheimer’s and Parkinson’s diseases (reviewed in Heneka et al., 2018). Multiple NLRP3 inhibitors are in clinical trials for a range of inflammatory diseases (Li et al., 2023). According to company information, selnoflast does not enter the brain.

NLRP3 is a receptor for Aβ, and mediates the innate immune response to amyloid in microglia, cells involved in Alzheimer’s pathogenesis (Halle et al., 2008). Deleting NLRP3 in the APP/PS1 mouse model diminishes Aβ deposition, synapse loss, and memory deficits (Heneka et al., 2013). Loss of NLRP3 also prevents tau tangle formation in human tau-expressing mice in response to injected Aβ (Ising et al., 2019).

No preclinical data is published for selnoflast, also known as RO7486967. It is one of a series of compounds related to the NLRP3 inhibitor MCC950, which was shown to block inflammasome activation, promote microglial clearance of Aβ, reduce Aβ accumulation, and improve cognitive function in APP/PS1 mice (Coll et al., 2015; Dempsey et al., 2017). MCC950 also prevented inflammasome activation by fibrillar α-synuclein, and led to less neuron loss and better dopaminergic signaling in Parkinson’s disease models (Gordon et al., 2018). The scientists behind these studies founded Inflazome, which developed and held the patent on MCC950 and related compounds.

Findings

From September 2019 to February 2020, Inflazome conducted Phase 1 single- and multiple-ascending-dose study of selnoflast in 64 healthy adults and patients with cryopyrin-associated periodic syndrome. CAPS is an autoimmune disease caused by gain-of-function mutations in the NLRP3 gene, and affects both peripheral organs and the central nervous system. According to a February 2020 press release, the drug was safe and tolerable, with dose-proportional pharmacokinetics and pharmacodynamic effects.

In October 2020, Roche acquired Inflazome. The company abandoned a planned study of another NLRP3 inhibitor inzomelid/emlenoflast, and began developing selnoflast.

Starting in November 2021, Roche sponsored a Phase 1b study in 19 patients with ulcerative colitis, treated with 450 mg or placebo, once daily for seven days. Serum drug concentrations were claimed to exceed those required for 90 percent NLRP3 inhibition throughout the dosing period. Target engagement was confirmed by the diminished production of IL-1β by blood cells following ex-vivo LPS stimulation. Treatment did not change plasma IL-18, nor did it alter IL-1-related gene expression in colon tissue biopsies. Selnoflast-treated participants had more adverse events than those on placebo. None were serious; headache and indigestion were most common. The investigators concluded the drug was safe and well-tolerated, but unlikely to affect inflammation in patients with ulcerative colitis (Klughammer et al., 2023).

In September 2022, the company began a Phase 1b trial in 72 people with early Parkinson’s disease. The study, at 20 centers in Europe and the U.S., compares 28 days of RO7486967 to placebo against primary outcomes of adverse events and suicidality. Secondary outcomes are pharmacokinetics, and brain neuroinflammation measured by binding of the 18 kDa translocator protein (TSPO) PET ligand [18F]-DPA-714. The trial is expected to finish in January 2025.

Roche is running additional Phase 1b trials for asthma and coronary artery disease. A trial for chronic obstructive pulmonary disease finished in June 2022.

For details of these trials, search for selnoflast, RO7486967, or IZD-334 on clinicaltrials.gov and the International Clinical Trials Registry.

Last Updated: 21 May 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Research Models

Lrrk2 KO Mouse

Synonyms: Lrrk2 knockout mouse, LRRK2−/−

Tools

Back to the Top

Species: Mouse
Genes: Lrrk2
Modification: Lrrk2: Knock-Out
Disease Relevance: Parkinson's Disease
Strain Name: B6.129X1(FVB)-Lrrk2tm1.1Cai/J

Modification Details:

Summary

This knock-out (KO) model was generated by deleting exon 2 of the Lrrk2 gene, which results in a premature stop codon in exon 3. Brain tissue from Lrrk2 KO mice does not express full-length LRRK2 protein. KO mice are viable, fertile, and do not exhibit any developmental or gross physical abnormalities (The Jackson Laboratory; Parisiadou et al., 2009).

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

  • Dopamine Deficiency
  • Non-Motor Impairment
  • α-synuclein Inclusions
  • Neuronal Loss

No Data

Neuronal Loss

Neuronal Loss No  differences between KO and wild-type mice up to 24 months of age in the number of tyrosine hydroxylase (TH)–positive cells in the substantia nigra pars compacta. No neurodegeneration markers observed in the striatum and cortex at 20 months. Cerebral cortex and dorsal (but not ventral) striatum volumes reduced at 12 months.

Dopamine Deficiency

Levels  of TH in the striatum are equal between genotypes in 18- to 24-month-old mice.

α-synuclein Inclusions

No abnormal accumulation of α-synuclein in the cell bodies of striatal neurons observed in 20-month-old KO mice.

Neuroinflammation

Striatal staining of GFAP, a marker of reactive astrocytosis, did not differ between control and KO mice, but cells positive for Iba1 staining, a marker of activated microglia, were moderately enlarged in the striatum of 20 -month-old KO mice. Cx3cr1 mRNA levels higher in KO mouse brains.

Mitochondrial Abnormalities

Adult (9 - to 23-week-old) Lrrk2 KO mice exhibit enhanced mitophagy in dopaminergic neurons of the substantia nigra pars compacta, as detected by an increase in the number of mitolysosomes.

Motor Impairment

Motor behavior is generally intact up to 18 months based on Rotarod and open field tests. However, some age-dependent effects are observed on the open field test: 12 -month-old mice traveled longer distances and had higher walking speeds versus controls, which was not apparent in 3- or 24-month-old mice. Older (24 months) mice had deficits in motor skill learning as measured by Rotarod.

Non-Motor Impairment

No differences were observed between KO and wild-type mice across 6 to 24 months of age on several behavioral tests, including the elevated plus maze for anxiety-like behavior, the buried treat test to measure hyposmia, the grip strength test for forelimb strength, or working memory as measured by spontaneous alternation.

Last Updated: 20 May 2024

COMMENTS / QUESTIONS

No Available Comments

Make a comment or submit a question

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

NT-0796

Tools

Back to the Top

Overview

Name: NT-0796
Synonyms: Propan-2-yl (2R)-2-{[(1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)- carbamoyl]oxy}-3-(pyrimidin-2-yl)propanoate
Therapy Type: Small Molecule (timeline)
Target Type: Inflammation (timeline)
Condition(s): Parkinson's Disease
U.S. FDA Status: Parkinson's Disease (Phase 1/2)
Company: NodThera

Background

NT-0796 is an oral, brain-penetrant inhibitor of inflammasomes that contain NLRP3, aka nod-like receptor family pyrin domain-containing protein 3. Inflammasomes are multiprotein, cytosolic complexes that function as sensors in the innate immune system. They are found in microglia in the brain, and in monocytes and macrophages outside the brain. Their activation by pathologic proteins and other stressors triggers production and secretion of the proinflammatory cytokines IL1-β and IL-18. Inflammasomes can induce cell death.

NLRP3-containing inflammasome activation is implicated in a growing number of neurodegenerative conditions where chronic inflammation plays a role, including Alzheimer’s and Parkinson’s diseases, as well as in obesity and cardiovascular disease (reviewed in Li et al., 2023).

In Alzheimer’s mouse models, inhibition of the NLRP3 inflammasome has been shown to reduce amyloid and tau pathology (Heneka et al., 2013; Ising et al., 2020).

NT-0796 inhibits NLRP3 inflammasome-mediated cytokine production in human blood with an IC50 of 6.8 nanomolar (Harrison et al., 2023). It enters the brain of mice with a blood-to-brain ratio of 0.79. NT-0796 is a prodrug that undergoes intracellular conversion to its active form in human immune cells. This activation does not occur in mice, which lack the necessary enzyme. A humanized mouse line was produced for pharmacokinetic and pharmacodynamic profiling of the compound (Smolak et al., 2024). In diet-induced obesity in these mice, NT-0796 caused weight loss as potently as did the GLP1 inhibitor semaglutide, and improved markers of cardiovascular risk (Thornton et al., 2024). It did not cause weight loss in non-obese mice.

Findings

From August 2021 to August 2022, NodThera ran a Phase 1 first-in-human study to assess safety and pharmacokinetics of NT-0796 in 76 healthy volunteers. Single ascending doses from 1 to 300 mg were given as a liquid formulation. Multiple MAD cohorts were planned based on results of the single-dose study, and included CSF sampling. Pharmacodynamic outcomes included inflammatory cytokines IL-1β, IL-18, IL-6, and TNFα in serum, and after ex vivo stimulation of blood cells from participants. In May 2022, the company reported the drug was safe after single dosing, and showed dose-proportional pharmacokinetics. Treatment lowered inflammatory cytokine levels in the ex vivo blood cell assay (press release). A September 2022 press release on the multiple-dose part of the study claimed brain penetration to levels higher than needed to achieve an anti-inflammatory effect, and a reduction in blood levels of the inflammatory biomarker C-reactive protein after treatment. No doses were specified, or data shown.

In 2023, the company began a Phase 1b/2a trial to measure the effect of NT-0796 on inflammatory and disease specific biomarkers in patients with Parkinson’s disease (press release). This trial does not appear in registries. According to NodThera, the first part of the study tested a new capsule formation in healthy volunteers, who received 150 mg twice a day for one week. A cohort with Parkinson’s disease received the same dose for four weeks. In a July 2023 press release, the company claimed significant reductions in inflammatory markers in CSF in four healthy volunteers after one week treatment. They also claimed a 13 percent reduction in CSF neurofilament light chain after one week.

Results of the completed trial were presented at the March 2024 AD/PD conference. The drug appeared safe. Adverse events were mainly mild, transient, and unrelated to drug. There were no serious adverse events. The capsule formulation extended the drug’s half-life compared to the liquid. CSF levels of drug in PD patients were two to three times higher than in elderly volunteers with the same plasma levels. After one or four weeks of treatment, patients had reductions in CSF IL-1, and IL-6, and a trend toward reduction in neurofilament light chain and soluble Trem2. Blood markers of peripheral inflammation were also reduced. Participants did not lose weight. The company is conducting long-term toxicology studies to facilitate a new trial.

In October 2023, NodThera began a Phase 1/2 trial to assess inflammation, weight loss, and safety in obese volunteers.

For details on the Phase 1 trial, see ANZCTR.org. For other trials, see clinicaltrials.gov.

Last Updated: 16 May 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

Lixisenatide

Tools

Back to the Top

Overview

Name: Lixisenatide
Synonyms: Adlyxin, Lyxumia
Therapy Type: Other
Target Type: Other (timeline)
Condition(s): Parkinson's Disease
U.S. FDA Status: Parkinson's Disease (Phase 2)
Company: Sanofi
Approved for: Type 2 diabetes

Background

Lixisenatide is an analog of the hormone glucagon-like peptide-1. GLP-1 stimulates the pancreas to release insulin in response to food intake. GLP-1 mimetics resensitize cells to insulin signaling and are used to treat Type 2 diabetes. Several GLP-1 mimetics are also approved for weight loss. The FDA approved Adlyxin for Type 2 diabetes in 2016, but Sanofi discontinued marketing it for this disease in 2023, citing business reasons. Lixisenatide is taken as a once-daily self-injection under the skin.

Lixisenatide is one of four GLP-1 mimetics being tested for Alzheimer’s or Parkinson’s diseases. It is a 44-amino acid peptide closely related to exenatide (Leon et al., 2017). Lixisenatide appears to cross the blood-brain barrier better than liraglutide and semaglutide (Hunter and Hölscher, 2012; Salameh et al., 2020).

The rationale of using GLP-1 receptor agonists for neurodegeneration stems from observations of insulin resistance in some cases of Alzheimer's disease (e.g. Talbot et al., 2012; Talbot, 2014). Type 2 diabetes raises the risk of Parkinson’s, and in some studies, the prevalence of Parkinson’s was lower in people with diabetes who were treated with GLP-1 receptor agonists compared to those taking other diabetes medications (Brauer et al., 2020; Svenningsson et al., 2016).

GLP-1 mimetics are neuroprotective in preclinical models of Alzheimer’s and Parkinson’s (e.g. Perry et al., 2002; Bertilsson et al., 2008; Li et al., 2010). Potential mechanisms of neuroprotection by GLP-1 mimetics include reduced inflammation (reviewed in Reich and Hölscher, 2022). 

Preclinically, lixisenatide reduced Aβ-induced impairments in spatial learning and memory in rats, and was neuroprotective in APP/PS1/tau mice (Cai et al., 2014; Cai et al., 2017Cai et al., 2018). In the APPswe/PS1Δe9 mouse line, it improved object recognition and synaptic plasticity, prevented synapse loss, reduced amyloid load and microglial activation, and increased neurogenesis, all at lower doses than liraglutide (McClean and Hölscher, 2014; Hölscher, 2014). In the MPTP toxin-induced mouse model of Parkinson's lixisenatide prevented motor impairment and dopamine neuron loss (Liu et al., 2015).

For a review of the development of GLP-1 class drugs for Parkinson’s and Alzheimer’s, see Hölscher, 2024.

Findings

In June 2018, French researchers started a Phase 2 study to evaluate the effect of lixisenatide in people with early Parkinson’s disease. The LixiPark trial enrolled 156 participants, with a target dose of 20 μg daily for one year, or a matched placebo, in addition to their usual Parkinson's medications. The primary outcome was motor function, as measured by the Movement Disorders Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) Part 3. Conducted at 20 sites in France, the trial ended in April 2021. Results are published (see Apr 2024 news on Meissner et al., 2024). After 12 months, the lixisenatide group had improved by 0.04 points on the primary outcome, compared to a decline of 3.04 points in the placebo group, a statistically significant difference. A difference favoring treatment remained after two months off drug. Secondary outcomes including non-motor symptoms, activities of daily living, and need for levodopa were not different between the groups. One third of participants had to limit their dose to 10 μg due to side effects, mainly nausea, as well as vomiting and acid reflex. A post hoc analysis suggested greater benefit for people under 60.

For details on the LixiPark trial, see clinicaltrials.gov.

Last Updated: 14 May 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

Further Reading

No Available Further Reading

Therapeutics

PhotoBioModulation

Tools

Back to the Top

Overview

Name: PhotoBioModulation
Synonyms: Low level light therapy, Low level laser therapy, Cold laser therapy, Transcranial infrared brain stimulation, Transcranial photobiomodulation, Transcranial near-infrared laser therapy (NILT), Transcranial low-level light therapy
Therapy Type: Procedural Intervention
Target Type: Inflammation (timeline), Unknown
Condition(s): Alzheimer's Disease, Mild Cognitive Impairment, Parkinson's Disease
U.S. FDA Status: Alzheimer's Disease (Not Regulated), Mild Cognitive Impairment (Not Regulated), Parkinson's Disease (Not Regulated)

Background

Photobiomodulation is a form of light therapy that exposes tissue to red or near-infrared (NIR) light. Low-energy lasers or light-emitting diodes are placed on or near the skin. Their light penetrates tissue, where endogenous chromophores absorb it, and cause physical and chemical changes in cells. This non-heating therapy employs wavelengths from ~600 to ~1100 nm. Near-infrared light, with its longer wavelengths, penetrates tissues more deeply than shorter wavelengths of red light. This treatment causes no notable side effects, and can be done at home.

Red or NIR light is claimed to promote cell health by enhancing mitochondrial energy production, and by increasing blood circulation. Absorption of NIR light by the mitochondrial enzyme cytochrome C oxidase and subsequent stimulation of the respiratory chain is the commonly claimed mechanism of action (reviewed in Hamblin, 2018). This therapy has been studied in animal models, where it is reported to increase ATP production, stimulate anti-inflammatory, anti-apoptotic, and antioxidant responses, promote neurogenesis and synaptogenesis, and improve cognition (reviewed in Salehpour et al., 2018). In healthy rats, photobiomodulation (PBM) improved the metabolic profile, spatial memory, and neuroinflammation markers in young and aged animals (Dos Santos Cardoso et al., 2021; Cardoso et al., 2022).

Literature in animal models of Alzheimer’s disease reports improved mitochondrial function, reduced Aβ plaques, tau tangles, inflammation, and oxidative stress, and lessening of cognitive deficits after transcranial PBM (e.g. De Taboada et al., 2011; reviewed in Su et al., 2023; Salehpour et al., 2021). Targeting abdomen, bone marrow, or lymph nodes with light therapy also improved outcomes in mouse models of AD and PD (Oron and Oron, 2016; Johnstone et al., 2014; Wu et al., 2022; Farfara et al., 2015). PBM is claimed to activate microglia, stimulate glymphatic clearance of Aβ, and improve gut flora (Salehpour et al., 2022; Chen et al., 2021; Liebert et al., 2019; Stepanov et al., 2022). In a negative study, transcranial PBM with 810 nm light had no benefit in the 5XFAD mouse model (Sipion et al., 2023).

For a recent review on preclinical and clinical development of PBM for AD and PD, see Shen et al., 2024.

Parameters of time, wavelength, power, and light source for PBM are under investigation (e.g. Spera et al. 2021; Joshi et al., 2024). In extracranial low-energy applications, only a tiny fraction of light penetrates the skull to reach the brain, although applying higher light intensity can increase penetration (Henderson and Morries, 2015). Human cadaver studies have shown near-infrared wavelengths can penetrate 40 mm through the scalp and skull into the brain (Tedford et al., 2015). Alternative approaches such as intracranial and intranasal delivery are being investigated to reach deeper brain structures (e.g., Salehpour et al., 2020).

Many different PBM devices are sold directly to consumers for home use. Some are FDA-approved for treating pain and inflammation, wound healing, promoting hair growth, and decreasing fat deposits; many are not. Hundreds of clinical trials are registered for a wide range of applications. A Phase 3 trial in stroke patients was terminated for futility (Hacke et al., 2014). More recently, a placebo-controlled trial found no effect on cognitive impairment in people with schizophrenia (Kheradmand et al., 2022). Evidence for cognitive improvement in healthy adults is mixed (Salehpour et al., 2019; Lee et al., 2023).

Findings

Case reports and pilot studies claim benefits of PBM in people with mild cognitive impairment or dementia. For example, one patient with dementia reversed cognitive and olfactory dysfunction after daily PBM therapy to the head, lower back, and intranasally (Salehpour et al., 2019). In MCI, a trend was claimed toward improved blood flow and cognition after eight weeks of red light to the main arteries supplying the brain (Baik et al., 2021). A single session of light therapy was claimed to acutely improve memory in adults with MCI (Chan et al., 2021). Among 42 women with MCI, five sessions of transcranial PBM reportedly improved MMSE scores and measures of attention over sham treatment (Papi et al, 2022). 

From 2010-2012, the Quietmind Foundation sponsored a pilot trial in 11 people with early and mid-stage dementia, to assess whether repeated scalp exposure to six minutes of 1,072 nm infrared stimulation daily for 28 days improves cognitive and behavioral functioning as indicated by normalization of EEG activity, increased cerebral oxygenation and performance on standardized neuropsychological measures. The sham-controlled study lists a sole outcome of ADAS-Cog. Results are published, claiming trends toward a benefit in some parts of the ADAS-Cog related to executive functioning, and improvements in EEG parameters and functional connectivity (Berman et al., 2017). A follow-up study with 100 participants began in October 2018. This trial combined PBM with neurofeedback, and ran for eight weeks. Sixty people were enrolled, and the results are published (Nizamutdinov et al., 2021). Treatment was associated with improvements in the MMSE and several parts of the neuropsychological test battery, sleep, anxiety, mood, and daily routine.

Several device makers have sponsored trials for treatment of mild cognitive impairment or dementia. The Vielight Neuro Gamma device consists of five LEDs placed on the scalp to target default mode network nodes, plus an intranasal LED to target the hippocampus. All LEDS deliver 810 nm light, at either 10 or 40 Hz frequency. Both frequencies have been shown to modulate EEG alpha, beta, and gamma waves in healthy adults (e.g. Zomorrodi et al., 2019). An early study in five people with AD used 10 Hz and reported significant improvement on the MMSE and the ADAS-Cog after 12 weeks of treatment. Caregivers reported that patients slept better, had fewer angry outbursts, and less anxiety and wandering (Saltmarche et al., 2017).

In May 2017, a single-center trial at University of California, San Francisco, began testing the Vielight device in 20 dementia patients. It compared 12 weeks of 40 Hz therapy, delivered for 20 minutes three days a week at home, to usual care, reporting improvements in the ADAS-Cog and neuropsychiatric symptoms, increased cerebral blood flow, and increased connectivity within the default mode network in the first eight treated patients (Chao, 2019). A follow-up trial, completed in January 2021, tested active versus sham 40 Hz treatment for 16 weeks in 14 AD patients, against a primary outcome of change in ADAS-Cog. Results posted on clinicaltrials.gov indicate no changes with treatment in cognition or biomarkers of Aβ42, tau, and neurofilament light.

In December 2017, Vielight began a study in 60 people with moderate to severe AD at two sites in Ontario, comparing 12 weeks of active to sham home treatment with 40 Hz therapy for 20 minutes daily, six days a week. The primary outcome was change in the Severe Impairment Battery score. The company claims sham treatment is indistinguishable from active, as their device produces no heat and no visible light. This study was completed in February 2022. No results have been made public.

In June 2019, Vielight began a pivotal trial of the 40 Hz device. This trial planned to enroll 228 participants with severe dementia, for active versus sham treatment against endpoints of SIB and ADCS-ADL. This trial was supposed to finish in May 2023, but was suspended due to slow recruitment.

In June 2019, an academic study in Florida and Arizona began to study whether PBM could improve age-related cognitive and mood changes in older healthy adults and those with PD. The study plans to enroll 135 participants to be randomized to 12 weeks of photobiomodulation combining lab-based cranial stimulation with the MedX health Console plus intranasal stimulation at home with the Vielight device. The primary outcome is change in performance on the Arena task, a virtual reality version of the Morris water maze (Parslow et al., 2004). Completion is anticipated in October 2024.

The same university groups began a preventive trial in August 2020, involving 168 older adults at risk for AD, defined by subjective cognitive complaints and a first-degree family history of Alzheimer's disease. Participants are randomized to 12 weeks of in-lab cranial plus at-home intranasal treatment, against the primary outcome of the ARENA score. Completion was expected in April 2024.

In April 2021, the TRAP-AD trial began testing transcranial PBM in 125 people with MCI or mild AD dementia (Iosifescu et al., 2023). This academic, multisite, sham-controlled study is assessing the effects of continuous 808 nm NIR light delivered bilaterally to the forehead for 11 minutes, in 24 sessions over eight weeks. The device is an investigational helmet made by LiteCure medical laser company. Target engagement is to be confirmed by changes in fMRI blood oxygen-level dependent (BOLD) signal after one treatment (Gaggi et al., 2024). RBANS total score after eight weeks serves as the primary outcome; secondary outcomes include RBANS and other cognitive tests out to three months. This study also uses MRS imaging to assess change in mitochondrial function, and tau-PET to relate treatment effects to baseline tau burden. Completion is expected in November 2025.

In March 2023, a pilot trial began at Unity Health Toronto testing the Vielight device in 20 people with mild cognitive impairment due to AD. The treatment regimen involves 20 minutes per day, six days a week, for six weeks, and includes a sham control. The primary outcomes are MMSE, and memory and executive function tests. The trial will assess quality of life, blood and MRI biomarkers, sleep, and neuropsychiatric and depressive symptoms. Completion was expected in March 2024.

From September 2018 to May 2020, the French company REGEnLIFE ran a feasibility trial of its RGn530 device, which features a helmet and abdominal belt that deliver 10 Hz pulses of NIR and red light from a laser and LEDs. The study randomized 53 people with mild to moderate AD to active or sham treatment, before being terminated due to COVID. Results are published (Blivet et al., 2022). No serious adverse events were recorded. Patients complied, with most completing the prescribed 40 treatment sessions of 25 minutes each over eight weeks. The investigators claimed trends toward improvement on some cognitive measures.

In July 2023, REGEnLIFE began recruiting for the pivotal LIGHT4Life study. This sham-controlled trial plans to enroll 108 people with mild to moderate AD in multiple centers in France. Treatment consists of six months of 20 minute in-clinic sessions, beginning at five per week, and gradually ramping down to two per week (Blivet et al., 2024). The primary outcome is ADAS-Cog; 34 secondary and exploratory outcomes include other cognitive and functional tests, safety, blood biomarkers of amyloid, inflammation, tau, and neurodegeneration, as well as analysis of fecal microbiota, and metabolomics. Primary completion is expected in May 2025.

REGEnLIFE is also running a pilot in 50 people with concussion. This follows on clinical studies of transcranial NIR in people with traumatic brain injury that claim improvements in executive function, learning, and memory (Naeser et al., 2014).

A pilot study of an NIR device in Iran reported safety and benefits in people with AD (Razzaghi et al., 2024); studies on other devices began in late 2023 in China and Japan.

PBM is also being tested for Parkinson’s disease (for review, see Bicknell et al., 2024). A sham-controlled study in people with PD suggested that red light targeting the substantia nigra could speed walking (Santos et al., 2019). In an open-label study of 12 people, Australian investigators claimed better mobility, cognition, dynamic balance, and fine motor skills after 12 weeks to one year of in-clinic and at-home PBM to the abdomen, neck, head, and nose (Liebert et al., 2021). The treatment induced changes in the gut microbiome (Bicknell et al., 2022). Some patients improved with at-home PBM to just the abdomen and neck (Liebert et al., 2022). A sham controlled pilot subsequently tested an Australian company’s transcranial device in 20 people with PD, finding a 12-week course of treatment to be safe and feasible (Herkes et al., 2023). Another study in Australia reported that four weeks of transcranial plus intraoral treatment did not change cognition measured by the MoCA, although some improvement was measured in writing and walking. A placebo effect was seen (Bullock-Saxton et al., 2021). An ongoing trial in France is testing endoventricular PBM using an implanted light source (see Darlot et al., 2016).

For more on photobiomodulation trials for AD, see clinicaltrials.gov.

For more on photobiomodulation trials for PD, see clinicaltrials.gov.

Last Updated: 09 May 2024

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