Clinical Trials Logo

Clinical Trial Details — Status: Recruiting

Administrative data

NCT number NCT05260177
Other study ID # ASIII
Secondary ID
Status Recruiting
Phase N/A
First received
Last updated
Start date September 20, 2022
Est. completion date December 1, 2026

Study information

Verified date January 2024
Source Zealand University Hospital
Contact Peter Høgh, MD, Phd
Phone 47322809
Email phh@regionsjaelland.dk
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

The ALZLIGHT STAGE III Study is a continuation of the ALZLIGHT Pilot - Study on Safety, Feasibility and Neural Activation of Non-Invasive Light Therapy System. As with the first two stages, this study will examine whether entrainment of 40 Hz neural oscillation by novel 40 Hz Invisible Spectral Flicker is a potential therapy for Alzheimer's Disease. In order to examine this, 62 patients with mild to moderate Alzheimer's Disease will be recruited. The patients will be exposed to the Non-Invasive Light Therapy System for 1 hour a day for 6 months. The effect will be measured by a combination of electroencephalography, cognitive testing, functional magnetic resonance imaging, magnetic resonance spectroscopy and actigraphy.


Description:

Recent studies in mouse models of Alzhimer's Disease (AD) have shown that exposure to 40 Hz stroboscopic light therapy for one hour a day, resulted in slowing disease progression and lead to multiple neuroprotective effects such as cognition and memory recovery, and even scavenged both tau and Aβ protein species. Hence, the 40 Hz stroboscopic light therapy has a considerable potential for treatment of humans. This study will utilize a novel way of masked light by alternating the spectral composition of a white light, rendering the flicker invisible to the conscience perception, while still entraining 40 Hz oscillations in the brain. In the study, 62 patients with probable mild to moderate AD will be exposed to either invisible spectral flickering light through the Light Therapy System (LTS) (active setting) or continuous non-flickering white light (sham setting) for 1 hour each day. The sham setting is a high quality sham intervention as subjects will be blinded to the setting, both appear as white light. The study will last 8½ months for each participant and consist of 3 periods: Enrollment period of 1 month, an intervention period of 6 months, and a 1½ month post-interventional follow-up period. In order to test whether the LTS intervention is a potential treatment for AD, cognition will be measured by neuropsychological tests at baseline and at follow-ups. To get a better understanding of the potential effects, markers of efficacy based on MRI, MRS, EEG and blood samples will be tested. The results from this study will increase the understanding of the impact of gamma oscillations in the human brain, and how it can be utilized as a novel and important tool for the treatment of neurodegenerative diseases.


Recruitment information / eligibility

Status Recruiting
Enrollment 62
Est. completion date December 1, 2026
Est. primary completion date May 30, 2025
Accepts healthy volunteers No
Gender All
Age group 55 Years and older
Eligibility Inclusion Criteria: - Adult competent person, able to understand the nature of the study and give written informed consent. - Diagnosed with probable mild to moderate AD based on NIA-AA diagnostic criteria. - Age > 55 years. Females must be post-menopausal. - Fluent in Danish. - > 8 years of normal school education - Pass a color-blindness test (Ishihara color test) - Have visual and auditory capabilities, and language skills necessary for neuropsychological testing. - Participants must have a designated caregiver, who is available to the participant and can provide the necessary assistance with using the LTS device and the Actigraph wearable at home and assist with clinical visits and other practical issues Exclusion Criteria: - Profound visual impairment (visual acuity > 0.5) provided correction with spectacles, if needed - Significant abnormalities related to important parts of the brain, e.g., the visual system, prefrontal cortex, or hippocampus, or relevant lesions detected by pre-trial imaging. - Prior history of significant diseases related to the visual system or the brain. - Medication: Use of any antiepileptic drugs, neuromodulating drugs or high dose of sedatives will be excluded. - Prior history of substance abuse within the past 2 years. - Any significant systemic illness or unstable medical condition, which could lead to difficulty complying with the protocol (at the discretion of the PI)

Study Design


Related Conditions & MeSH terms


Intervention

Device:
Light Therapy System (LTS): Active Setting
Exposure for 1 hour á day for consecutive days
Light Therapy System (LTS): Sham Setting
Exposure for 1 hour á day for consecutive days

Locations

Country Name City State
Denmark Zealand University Hospital Roskilde

Sponsors (5)

Lead Sponsor Collaborator
Zealand University Hospital Göteborg University, OptoCeutics, Technical University of Denmark, University of Copenhagen

Country where clinical trial is conducted

Denmark, 

References & Publications (8)

Adaikkan C, Middleton SJ, Marco A, Pao PC, Mathys H, Kim DN, Gao F, Young JZ, Suk HJ, Boyden ES, McHugh TJ, Tsai LH. Gamma Entrainment Binds Higher-Order Brain Regions and Offers Neuroprotection. Neuron. 2019 Jun 5;102(5):929-943.e8. doi: 10.1016/j.neuron.2019.04.011. Epub 2019 May 7. — View Citation

Adaikkan C, Tsai LH. Gamma Entrainment: Impact on Neurocircuits, Glia, and Therapeutic Opportunities. Trends Neurosci. 2020 Jan;43(1):24-41. doi: 10.1016/j.tins.2019.11.001. Epub 2019 Dec 10. — View Citation

Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simren J, Montoliu-Gaya L, Pannee J, O Connor A, Weston PSJ, Lantero-Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med. 2021 Sep;290(3):583-601. doi: 10.1111/joim.13332. Epub 2021 Jun 26. — View Citation

Benedet AL, Mila-Aloma M, Vrillon A, Ashton NJ, Pascoal TA, Lussier F, Karikari TK, Hourregue C, Cognat E, Dumurgier J, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, Salvado G, Shekari M, Operto G, Gispert JD, Minguillon C, Fauria K, Kollmorgen G, Suridjan I, Zimmer ER, Zetterberg H, Molinuevo JL, Paquet C, Rosa-Neto P, Blennow K, Suarez-Calvet M; Translational Biomarkers in Aging and Dementia (TRIAD) study, Alzheimer's and Families (ALFA) study, and BioCogBank Paris Lariboisiere cohort. Differences Between Plasma and Cerebrospinal Fluid Glial Fibrillary Acidic Protein Levels Across the Alzheimer Disease Continuum. JAMA Neurol. 2021 Dec 1;78(12):1471-1483. doi: 10.1001/jamaneurol.2021.3671. — View Citation

Herrmann CS. Human EEG responses to 1-100 Hz flicker: resonance phenomena in visual cortex and their potential correlation to cognitive phenomena. Exp Brain Res. 2001 Apr;137(3-4):346-53. doi: 10.1007/s002210100682. — View Citation

Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, Mathys H, Seo J, Kritskiy O, Abdurrob F, Adaikkan C, Canter RG, Rueda R, Brown EN, Boyden ES, Tsai LH. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature. 2016 Dec 7;540(7632):230-235. doi: 10.1038/nature20587. Erratum In: Nature. 2018 Oct;562(7725):E1. — View Citation

Kasteleijn-Nolst Trenite D, Rubboli G, Hirsch E, Martins da Silva A, Seri S, Wilkins A, Parra J, Covanis A, Elia M, Capovilla G, Stephani U, Harding G. Methodology of photic stimulation revisited: updated European algorithm for visual stimulation in the EEG laboratory. Epilepsia. 2012 Jan;53(1):16-24. doi: 10.1111/j.1528-1167.2011.03319.x. Epub 2011 Nov 16. — View Citation

Martorell AJ, Paulson AL, Suk HJ, Abdurrob F, Drummond GT, Guan W, Young JZ, Kim DN, Kritskiy O, Barker SJ, Mangena V, Prince SM, Brown EN, Chung K, Boyden ES, Singer AC, Tsai LH. Multi-sensory Gamma Stimulation Ameliorates Alzheimer's-Associated Pathology and Improves Cognition. Cell. 2019 Apr 4;177(2):256-271.e22. doi: 10.1016/j.cell.2019.02.014. Epub 2019 Mar 14. — View Citation

Outcome

Type Measure Description Time frame Safety issue
Primary Gamma oscillations assessment Determine the total gamma power at 40 Hz, with no concomitant LTS device stimulation, assess changes in the gamma power at 40 Hz. Change from Baseline to 6 months and 7.5 months
Primary Induction of 40 Hz Gamma oscillations Estimate the change in electrical field patterns by EEG SSVEP, assess the difference between placebo and treatment for power spectral density signal to noise ratio at baseline measured by EEG SSVEP Change from Baseline to 6 months and 7.5 months
Secondary Cognition and memory assessment Assess changes in cognition measured by the Alzheimer's Disease Assessment Scale -Cognitive Subscale plus Executive Functioning and Functional Ability (ADAS Cog plus EF & FA). The score ranges from 0 to 135. A higher score reflects greater cognitive impairment. Change from Baseline to 6 months and 7.5 months
Secondary Cognition and memory assessment Assess changes in cognition measured by the Alzheimer's Disease Cooperative Study - Activities of Daily Living Inventory (ADCS-ADL). The score ranges from 0 to 78. A higher score reflects a better outcome. Change from Baseline to 6 months and 7.5 months
Secondary Cognition and memory assessment Assess changes in cognition measured by the Montreal Cognitive Assessment (MoCA). The score ranges from 0 to 30. A higher score reflects a better outcome. Change from Baseline to 6 months and 7.5 months
Secondary Connectivity measures rs-fMRI Connectivity: Estimate the temporal correlation between cortical regions, assess changes in correlation between cortical regions from baseline to 6 months and 7.5 months Change from Baseline to 6 months and 7.5 months
Secondary Connectivity measures EEG Connectivity: Estimate the temporal correlation between cortical regions, assess changes in correlation between cortical regions from baseline to 6 months and 7.5 months Change from Baseline to 6 months and 7.5 months
Secondary MR Spectroscopy MR Spectroscopy biomarkers: Assess changes from baseline to 6 months and 7.5 months Change from Baseline to 6 months and 7.5 months
Secondary Sleep Quality Assess changes from baseline to 6 months and 7.5 months of total sleep time in minutes, measured by actigraphy data and self-reported sleeping patterns (self-reported sleep quality scores based on patient's subjective report alone are often inaccurate).
Unit: total sleep time in minutes
Change from Baseline to 6 months and 7.5 months
Secondary Sleep Quality Assess changes from baseline to 6 months and 7.5 months of wakefulness after sleep onset, measured in minutes of wakefulness after a patient has fallen asleep based on actigraphy data.
Unit: total time of wakefulness in minutes
Change from Baseline to 6 months and 7.5 months
Secondary Biomarkers of Alzheimer's Disease Assess changes in biomarkers of Alzheimer's Disease in blood sampled from the participants from baseline to 6 months and 7.5 months. Markers of AD will be measured via ultrasensitive assays using fluid-based biomarkers such as plasma levels associated with amyloid pathology (plasma Aß42/40 ratio), tau (plasma P-tau181 and P-tau231), neurodegeneration (plasma neurofilament light), and astrocytic function (glial fibrillary acidic protein). Change from Baseline to 6 months and 7.5 months
Secondary Safety Assessment Estimate the safety of the LTS therapy, assess device- and procedure-related adverse events (DR/PR-AEs) including serious AEs (SAEs) occuring at any time during the trial 9 months
Secondary Feasibility assessment Investigate whether participants can meet the requirements of sitting in front of the LTS device for 1 hour per day. The feasibility of the LTS intervention will be measured by the amount of time (in minutes) of correct device use per day and through a self-report of usage via a compliance/feasibility questionnaire (structured interview on participant's self-reported usage and perception of the LTS device).
Unit: minutes per day of usage
Baseline to 6 months
Secondary Compliance assessment Investigate the tolerability of the LTS intervention through questionnaires (structured interviews) measured by the number of protocol breaches in total, i.e., not complying with one hour of light stimulation per day during the intervention period, and qualitative assessment based on the compliance/feasibility questionnaire (structured interview on participant's self-reported usage and perception of the LTS device).
Unit: number of total protocol breaches
Baseline to 6 months
Secondary MRI Atrophy assessment Assess changes from baseline to 6 months and 7.5 months of global atrophy (ventricular volume and hippocampal volume) using advanced MR techniques on structural MRI data, i.e., including but not limited to voxel-based analysis. Change from Baseline to 6 months and 7.5 months
Secondary MRI perfusion assessment Assess MRI perfusion: Changes from baseline to 6 months and 7.5 months. Change from Baseline to 6 months and 7.5 months
Secondary EEG: Spectral feature assessment Assess spectral features via rs-EEG Fourier power. Change from Baseline to 6 months and 7.5 months
See also
  Status Clinical Trial Phase
Completed NCT05040048 - Taxonomy of Neurodegenerative Diseases : Observational Study in Alzheimer's Disease and Parkinson's Disease
Withdrawn NCT03316898 - A FDG-PET Study of AGN-242071 Added to Standard-of-Care (Donepezil ± Memantine) for the Treatment of Participants With Mild to Moderate Alzheimer's Disease Phase 1
Withdrawn NCT02860065 - CPC-201 Alzheimer's Disease Type Dementia: PET Study Phase 2
Completed NCT01315639 - New Biomarker for Alzheimer's Disease Diagnostic N/A
Not yet recruiting NCT03740178 - Multiple Dose Trial of MK-4334 in Participants With Alzheimer's Clinical Syndrome (MK-4334-005) Phase 1
Recruiting NCT05607615 - A 6-Month Study to Evaluate the Safety & Potential Efficacy of Trappsol Cyclo in Patients With Early Alzheimer's Disease Phase 2
Terminated NCT03307993 - Positron Emission Tomography (PET) Study in Patients With Alzheimer's Disease Phase 1
Recruiting NCT02912936 - A Medium Chain Triglyceride Intervention for Patients With Alzheimer Disease N/A
Active, not recruiting NCT02899091 - Evaluation of the Safety and Potential Therapeutic Effects of CB-AC-02 in Patients With Alzheimer's Disease Phase 1/Phase 2
Not yet recruiting NCT02868905 - Identification of Epigenetic Markers Common to Obesity and Alzheimer's Disease in Women N/A
Completed NCT02907567 - Clinical Trial of CT1812 in Mild to Moderate Alzheimer's Disease Phase 1/Phase 2
Completed NCT02580305 - SUVN-502 With Donepezil and Memantine for the Treatment of Moderate Alzheimer's Disease- Phase 2a Study Phase 2
Completed NCT02516046 - 18F-AV-1451 Autopsy Study Phase 3
Terminated NCT02521558 - Effectiveness of Home-based Electronic Cognitive Therapy in Alzheimer's Disease N/A
Recruiting NCT02247180 - Cognitive Rehabilitation in Alzheimer`s Disease N/A
Terminated NCT02220738 - Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of ABT-957 in Subjects With Mild-to-Moderate Alzheimer's Disease on Stable Doses of Acetylcholinesterase Inhibitors Phase 1
Completed NCT02256306 - The PLasma for Alzheimer SymptoM Amelioration (PLASMA) Study N/A
Completed NCT02260167 - Treatment of Alzheimer's and Dementia With the Metabolism, Infections, Nutrition, Drug Elimination (MIND) Protocol N/A
Completed NCT02317523 - Alzheimer's Caregiver Coping: Mental and Physical Health N/A
Completed NCT01920672 - Disrupted Sleep, Neuroendocrine Status and the Behavioral Symptoms of AD N/A