search
Back to results

Study on the Effect of 40 Hz Non-Invasive Light Therapy System

Primary Purpose

Alzheimer's Disease

Status
Recruiting
Phase
Not Applicable
Locations
Denmark
Study Type
Interventional
Intervention
Light Therapy System (LTS): Active Setting
Light Therapy System (LTS): Sham Setting
Sponsored by
Zealand University Hospital
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Alzheimer's Disease focused on measuring Gamma Entrainment, 40 Hz, Invisible Spectral Flicker, LED, Light Therapy, Brain Stimulation, Gamma Oscillations, Gamma Induction

Eligibility Criteria

55 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

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)

Sites / Locations

  • Zealand University HospitalRecruiting

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Sham Comparator

Arm Label

Active

Sham

Arm Description

Exposure to LTS device set to 40 Hz invisible spectral flicker for 1 hour a day for consecutive days

Exposure to LTS device set to continuous color matched white light for 1 hour a day for consecutive days

Outcomes

Primary Outcome Measures

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.
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

Secondary Outcome Measures

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.
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.
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.
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
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
MR Spectroscopy
MR Spectroscopy biomarkers: Assess changes from baseline to 6 months and 7.5 months
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
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
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 Aβ42/40 ratio), tau (plasma P-tau181 and P-tau231), neurodegeneration (plasma neurofilament light), and astrocytic function (glial fibrillary acidic protein).
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
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
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
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.
MRI perfusion assessment
Assess MRI perfusion: Changes from baseline to 6 months and 7.5 months.
EEG: Spectral feature assessment
Assess spectral features via rs-EEG Fourier power.

Full Information

First Posted
January 26, 2022
Last Updated
December 12, 2022
Sponsor
Zealand University Hospital
Collaborators
OptoCeutics, University of Copenhagen, Technical University of Denmark, Göteborg University
search

1. Study Identification

Unique Protocol Identification Number
NCT05260177
Brief Title
Study on the Effect of 40 Hz Non-Invasive Light Therapy System
Official Title
ALZLIGHT Stage III - Study on the Effect of 40 Hz Non-Invasive Light Therapy System
Study Type
Interventional

2. Study Status

Record Verification Date
December 2022
Overall Recruitment Status
Recruiting
Study Start Date
September 20, 2022 (Actual)
Primary Completion Date
June 30, 2023 (Anticipated)
Study Completion Date
May 31, 2025 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Zealand University Hospital
Collaborators
OptoCeutics, University of Copenhagen, Technical University of Denmark, Göteborg University

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief 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.
Detailed 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.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Alzheimer's Disease
Keywords
Gamma Entrainment, 40 Hz, Invisible Spectral Flicker, LED, Light Therapy, Brain Stimulation, Gamma Oscillations, Gamma Induction

7. Study Design

Primary Purpose
Treatment
Study Phase
Not Applicable
Interventional Study Model
Parallel Assignment
Model Description
A parallel-group randomized (1:1), double-blinded, placebo-controlled, clinical trial
Masking
ParticipantInvestigatorOutcomes Assessor
Masking Description
Double-blinded
Allocation
Randomized
Enrollment
62 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Active
Arm Type
Experimental
Arm Description
Exposure to LTS device set to 40 Hz invisible spectral flicker for 1 hour a day for consecutive days
Arm Title
Sham
Arm Type
Sham Comparator
Arm Description
Exposure to LTS device set to continuous color matched white light for 1 hour a day for consecutive days
Intervention Type
Device
Intervention Name(s)
Light Therapy System (LTS): Active Setting
Intervention Description
Exposure for 1 hour á day for consecutive days
Intervention Type
Device
Intervention Name(s)
Light Therapy System (LTS): Sham Setting
Intervention Description
Exposure for 1 hour á day for consecutive days
Primary Outcome Measure Information:
Title
Gamma oscillations assessment
Description
Determine the total gamma power at 40 Hz, with no concomitant LTS device stimulation, assess changes in the gamma power at 40 Hz.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Induction of 40 Hz Gamma oscillations
Description
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
Time Frame
Change from Baseline to 6 months and 7.5 months
Secondary Outcome Measure Information:
Title
Cognition and memory assessment
Description
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.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Cognition and memory assessment
Description
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.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Cognition and memory assessment
Description
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.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Connectivity measures
Description
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
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Connectivity measures
Description
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
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
MR Spectroscopy
Description
MR Spectroscopy biomarkers: Assess changes from baseline to 6 months and 7.5 months
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Sleep Quality
Description
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
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Sleep Quality
Description
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
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Biomarkers of Alzheimer's Disease
Description
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 Aβ42/40 ratio), tau (plasma P-tau181 and P-tau231), neurodegeneration (plasma neurofilament light), and astrocytic function (glial fibrillary acidic protein).
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
Safety Assessment
Description
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
Time Frame
9 months
Title
Feasibility assessment
Description
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
Time Frame
Baseline to 6 months
Title
Compliance assessment
Description
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
Time Frame
Baseline to 6 months
Title
MRI Atrophy assessment
Description
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.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
MRI perfusion assessment
Description
Assess MRI perfusion: Changes from baseline to 6 months and 7.5 months.
Time Frame
Change from Baseline to 6 months and 7.5 months
Title
EEG: Spectral feature assessment
Description
Assess spectral features via rs-EEG Fourier power.
Time Frame
Change from Baseline to 6 months and 7.5 months

10. Eligibility

Sex
All
Minimum Age & Unit of Time
55 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
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)
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Peter Høgh, MD, Phd
Phone
47322809
Email
phh@regionsjaelland.dk
First Name & Middle Initial & Last Name or Official Title & Degree
Maibritt Horning, MSc
Phone
+45 81949649
Email
maibho@regionsjaelland.dk
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Maibritt Horning, MSc
Organizational Affiliation
Zealand University Hospital, Department of Neurology
Official's Role
Study Chair
First Name & Middle Initial & Last Name & Degree
Mikkel Pejstrup Agger, MD
Organizational Affiliation
Zealand Univeristy Hospital, Department of Neurology
Official's Role
Study Chair
First Name & Middle Initial & Last Name & Degree
Peter Høgh, MD, Phd
Organizational Affiliation
Zealand Univeristy Hospital, Department of Neurology
Official's Role
Principal Investigator
Facility Information:
Facility Name
Zealand University Hospital
City
Roskilde
ZIP/Postal Code
4000
Country
Denmark
Individual Site Status
Recruiting
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Peter Høgh, MD, Phd
Phone
47322809
Email
phh@regionsjaelland.dk
First Name & Middle Initial & Last Name & Degree
Maibritt Horning, MSc
Phone
+4581949649
Email
maibho@regionsjaelland.dk
First Name & Middle Initial & Last Name & Degree
Peter Høgh, MD, Phd
First Name & Middle Initial & Last Name & Degree
Maibritt Horning, MSc
First Name & Middle Initial & Last Name & Degree
Mikkel Pejstrup Agger, MD

12. IPD Sharing Statement

Plan to Share IPD
No
Citations:
PubMed Identifier
22091642
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.
Results Reference
background
PubMed Identifier
11355381
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.
Results Reference
background
PubMed Identifier
30879788
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.
Results Reference
background
PubMed Identifier
27929004
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.
Results Reference
background
PubMed Identifier
31836315
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.
Results Reference
background
PubMed Identifier
31076275
Citation
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.
Results Reference
background
PubMed Identifier
34021943
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.
Results Reference
background
PubMed Identifier
34661615
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.
Results Reference
background
Links:
URL
https://doi.org/10.1117/12.2544338
Description
Carstensen M et al. 40 Hz invisible spectral flicker and its potential use in Alzheimer's light therapy treatment. Proc. SPIE 11221, Mechanisms of Photobiomodulation Therapy XV, 112210L (11 March 2020)

Learn more about this trial

Study on the Effect of 40 Hz Non-Invasive Light Therapy System

We'll reach out to this number within 24 hrs