WO2024064897A1 - Methods of reducing neurodegeneration associated with neurodegenerative diseases - Google Patents
Methods of reducing neurodegeneration associated with neurodegenerative diseases Download PDFInfo
- Publication number
- WO2024064897A1 WO2024064897A1 PCT/US2023/074913 US2023074913W WO2024064897A1 WO 2024064897 A1 WO2024064897 A1 WO 2024064897A1 US 2023074913 W US2023074913 W US 2023074913W WO 2024064897 A1 WO2024064897 A1 WO 2024064897A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- subject
- tau
- measurement
- lemborexant
- dose
- Prior art date
Links
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6893—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
- G01N33/6896—Neurological disorders, e.g. Alzheimer's disease
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/506—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/28—Neurological disorders
- G01N2800/2814—Dementia; Cognitive disorders
- G01N2800/2821—Alzheimer
Definitions
- AD Alzheimer’s disease
- AD is a progressive, neurodegenerative disorder of unknown etiology and the most common form of dementia among older people.
- there were 26.6 million cases of AD in the world (range: 11.4-59.4 million) (Brookmeyer, R., et al., Forecasting the global burden of Alzheimer’s Disease. Alzheimer Dement.2007; 3:186-91), while there were more than 5 million people in the United States reportedly living with AD (Alzheimer’s Association, Alzheimer’s Association report, 2010 Alzheimer’s disease facts and figures. Alzheimer Dement.2010; 6:158-94).
- AD Alzheimer disease
- AD Alzheimer's disease
- a ⁇ amyloid beta peptide
- a ⁇ exists in various conformational states - monomers, oligomers, protofibrils, and insoluble fibrils. Details of the mechanistic relationship between onset of Alzheimer’s disease and A ⁇ production is unknown. However, some anti-A ⁇ antibodies are undergoing clinical study now as potential therapeutic agents for Alzheimer’s disease. [007] In addition to neuritic plaques, the disease is also characterized by tau aggregation and hyperphosphorylation, increased immune response, neuronal degeneration, synaptic loss, and eventual cognitive dysfunction, dementia, and death. Attorney Docket No.: 08061.0057-00304 [008] Insomnia has been implicated as a risk factor for AD.
- insomnia has been treated with various medicaments, including doxepin, a tricyclic antidepressant (TCA), and dual orexin receptor antagonists (also called DORAs) such as suvorexant or lemborexant.
- TCA tricyclic antidepressant
- DORAs dual orexin receptor antagonists
- Lemborexant and methods of using it are disclosed in, e.g., U.S. Patent Nos.11,026,944 and 11,096,941, the contents of which are herein incorporated by reference.
- Lemborexant has the following structure: and is also known as (1R,2S)-2-(((2,4-dimethylpyrimidin-5-yl)oxy)methyl)-2-(3- fluorophenyl)-N-(5-fluoropyridin-2-yl)cyclopropanecarboxamide or (1R,2S)-2-(((2,4- dimethylpyrimidin-5-yl)oxy)methyl)-2-(3-fluorophenyl)-N-(5-fluoropyridin-2- yl)cyclopropane-1-carboxamide.
- AD Alzheimer’s disease
- a pharmaceutically acceptable salt thereof or a solvate thereof
- treating AD comprises reducing and/or slowing cognitive decline.
- treating AD comprises affecting a change (e.g., slowing, delaying, or reducing) in at least one marker of AD pathology.
- the marker is a level of phosphorylation of tau, neurodegeneration, a change in microglial response, and/or presence of A ⁇ plaques.
- the marker is present in a brain region in the subject.
- the brain region is the hippocampus, somatomotor cortex, somatosensory cortex, piriform cortex, and/or entrorhinal cortex.
- the marker is detected in a body fluid of the subject.
- the body fluid is blood or cerebrospinal fluid (CSF).
- the subject does not show signs of dementia and/or cognitive impairment. In some embodiments, the subject has mild cognitive impairment or mild dementia.
- the subject is amyloid positive. In some embodiments, the subject is at risk for further A ⁇ accumulation. In some embodiments, the subject is an ApoE4 carrier.
- the subject Attorney Docket No.: 08061.0057-00304 has intermediate levels of amyloid PET (e.g., 20-40 centiloids). In some embodiments, the subject has elevated levels of amyloid PET (e.g., > 40 centiloids).
- the subject has been diagnosed with AD, based on brain imaging, cognitive function, and/or biomarker criteria.
- the subject has early AD. In some embodiments, the subject has pre- AD.
- An aspect of the present disclosure relates to a method of reducing or maintaining tau (e.g., reducing or maintaining a level of tau relative to the level before the start of treatment, or delaying tau accumulation, tau phosphorylation, and/or tau spreading, or slowing a rate of any of these) in a subject having AD or at risk of developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, wherein the therapeutically effective amount is sufficient to reduce or maintain tau in the subject.
- the subject is amyloid negative.
- the tau levels are reduced or maintained relative to a reference.
- the method comprises reducing and/or delaying tau accumulation and/or tau spreading, and/or slowing a rate thereof, as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference is a measurement from a control subject administered a placebo.
- the method comprises altering tau in a brain region of the subject.
- the method comprises altering the tau PET signal in a brain region of the subject.
- the brain region is the hippocampus, entorhinal cortex, and/or the piriform cortex.
- the method comprises reducing tau in a body fluid of the subject.
- the body fluid is blood or CSF.
- the tau is total tau.
- the tau is an insoluble tau.
- the tau is aggregated tau.
- the tau is a phosphorylated form of tau (phospho-tau).
- the phopho-tau is phosphorylated on one or more of T181, T217, S202, S205, or T231.
- the method comprises altering a ratio of phopho-tau to total tau.
- the ratio of phospho-tau to total tau is reduced compared to the ratio of CSF phospho-tau to total tau of the subject prior to administration of lemborexant. In some embodiments, the ratio of phospho-tau to total tau is maintained within 10% of the ratio of phospho-tau to total tau of the subject prior to the administration of lemborexant. In some embodiments, the method comprises increasing a rate of dephosphorylation of phospho-tau. In some embodiments, the method comprises reducing a rate of phosphorylation of tau. In some embodiments, the method comprises reducing or maintaining tau within 48 hours of administration of a first dose of lemborexant.
- the method comprises reducing phospho-tau in the hippocampus, entorhinal cortex, and/or piriform cortex.
- Another aspect of the present disclosure relates to a method of altering neurodegeneration (e.g., reducing and/or delaying neurodegeneration, and/or slowing a rate thereof) in a subject having AD or at risk of developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, Attorney Docket No.: 08061.0057-00304 wherein the therapeutically effective amount is sufficient to alter neurodegeneration in the subject.
- the subject is amyloid negative.
- altering neurodegeneration comprises reducing and/or delaying neurodegeneration, and/or slowing a rate thereof, as compared to a reference.
- the neurodegeneration is altered relative to a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference is a measurement from a control subject administered a placebo.
- the neurodegeneration is characterized by a loss of at least one of cortical thickness and hippocampal volume.
- altering neurodegeneration comprises maintaining or slowing a loss reduction of cortical thickness and/or hippocampal volume.
- the neurodegeneration is characterized by a loss of at least one of pyramidal neurons in the cortex, pyramidal neurons in the hippocampus, or granule cells in the hippocampus.
- altering neurodegeneration comprises maintaining or reducing loss of pyramidal neurons and/or granule cells.
- altering neurodegeneration comprises reducing a rate of neurodegeneration.
- altering neurodegeneration comprises altering a neurofilament light chain (NfL) level.
- the method comprises altering the NfL levels in the blood and/or CSF of the subject.
- a further aspect of the present disclosure relates to a method of altering A ⁇ plaques (e.g., reducing or delaying formation of A ⁇ plaques, or slowing a rate of growth thereof) in a subject having AD or at risk of developing AD, comprising Attorney Docket No.: 08061.0057-00304 administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, wherein the therapeutically effective amount is sufficient to alter A ⁇ plaques in the subject.
- the A ⁇ plaques are altered relative to a reference.
- altering A ⁇ plaques comprises reducing and/or delaying formation of A ⁇ plaques, and/or slowing a rate thereof, as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference is measurement from a control subject administered a placebo.
- the A ⁇ plaques are fibrillar plaques.
- the A ⁇ plaques all plaques (e.g., including diffuse plaques).
- altering A ⁇ plaques comprises reducing the growth of A ⁇ plaques.
- the method comprises reducing growth of A ⁇ plaques in the hippocampus of the subject, somatomotor cortex, the somatosensory cortex, and/or the piriform cortex of the subject.
- altering A ⁇ plaques comprises altering an amyloid PET signal obtained from a brain region of the subject.
- altering A ⁇ plaques corresponds to a reduction in the concentration of A ⁇ in the subject’s CSF.
- the A ⁇ is A ⁇ 38, A ⁇ 40, and/or A ⁇ 42.
- altering A ⁇ plaques within 48 hours of administration of a first dose of lemborexant.
- the subject does not show signs of dementia and/or cognitive impairment. In some embodiments, the subject has mild cognitive impairment or mild dementia. [032] In some embodiments, the subject is at risk for further A ⁇ accumulation. In some embodiments, the subject is an ApoE4 carrier. In some embodiments, the subject has intermediate levels of amyloid PET (e.g., 20-40 centiloids). In some embodiments, the subject has elevated levels of amyloid PET (e.g., > 40 centiloids). [033] In some embodiments, the subject has early-stage AD. In some embodiments, the subject has pre-AD.
- One aspect of the present disclosure relates to a method of modulating a microglial response in a subject having Alzheimer’s disease (AD) or at risk of developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, wherein the therapeutically effective amount is sufficient to modulate the microglial response in the subject.
- modulating the microglial response comprises modulating expression of at least one microglial marker.
- the microglial marker is a general microglial marker.
- the general microglial marker is Iba1, Clec7a, or CD68.
- the microglial marker is a homeostatic microglial marker.
- the homeostatic microglial marker is TMEM119 or P2RY12.
- modulating the microglial response comprises modulating activity of phagocytic microglia.
- Attorney Docket No.: 08061.0057-00304 [036]
- the subject has mild cognitive impairment or mild dementia.
- the subject does not show signs of dementia and/or cognitive impairment.
- the subject is amyloid negative.
- the subject has tau pathology.
- the subject has neurodegeneration in a brain region.
- the brain region is the hippocampus, the entorhinal cortex, and/or the piriform cortex. In some embodiments, the brain region is the CA1 region, the CA2 region, the CA3 region, or the dentate gyrus in the hippocampus.
- modulating the microglial response comprises modulating a response in microglia associated with degenerating neurons. In some embodiments, modulating the microglial response comprises reducing expression of at least one general microglial marker. In some embodiments, the general microglial marker is Iba1, CD68, or Clec7a. In some embodiments, modulating the microglial response comprises increasing expression of at least one homeostatic microglial marker.
- the homeostatic microglial marker is TMEM119 or P2RY12.
- the subject has A ⁇ plaques.
- the A ⁇ plaques are fibrillar A ⁇ plaques.
- the subject is at risk for further A ⁇ accumulation.
- the subject is an ApoE4 carrier.
- the subject has intermediate levels of amyloid PET (e.g., 20-40 centiloids).
- the subject has elevated levels of amyloid PET (e.g., > 40 centiloids).
- the subject has early-stage AD. In some embodiments, the subject has pre-AD.
- the A ⁇ plaques are present in the hippocampus, the somatomotor cortex, the somatosensory cortex, and/or the piriform cortex.
- modulating the microglial response comprises modulating a response in microglia associated with A ⁇ plaques.
- modulating the microglial response comprises increasing expression of a general microglial marker.
- the general microglial marker is Iba1, Clec7a, or CD68.
- modulating microglial response comprises increasing phagocytosis of A ⁇ plaques by phagocytic microglia.
- modulating the microglial response comprises reducing expression of a homeostatic microglial marker.
- the homeostatic microglial marker is TMEM119 or P2RY12.
- the therapeutically effective amount of lemborexant administered to the subject is in a range of 5 mg to 50 mg per day. In some embodiments, the therapeutically effective amount of lemborexant administered to the subject is in a range of 10 mg to 30 mg per day.
- the therapeutically effective amount of lemborexant administered to the subject is selected from 5 mg, 7.5 mg, 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg and 30 mg per day. [045] In some embodiments, the therapeutically effective amount of lemborexant administered to the subject is 20-25 mg per day. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day. [046] In some embodiments, lemborexant is administered at a first dose for a first period, a second dose for a second period, and optionally, at a third dose for a Attorney Docket No.: 08061.0057-00304 third period.
- each of the first period, the second period, and the third period are 1 week.
- the first dose is lower than the second dose, and optionally, the second dose is lower than the third dose.
- the first dose is 5 mg of lemborexant once per day
- the second dose is 10 mg of lemborexant once per day
- the third dose is 20-25 mg of lemborexant once per day.
- the first dose is 5 mg or 7.5 mg lemborexant once per day
- the second dose is 10 mg, 12.5 mg, 15 mg, or 17.5 mg of lemborexant once per day
- the third dose is 20 mg, 22.5 mg, 25 mg, 27.5 mg or 30 mg of lemborexant once per day.
- the first dose is higher than the second dose, and optionally, the second dose is higher than the third dose.
- the first dose is 20-25 mg of lemborexant once per day
- the second dose is 10 mg of lemborexant once per day
- the third dose is 5 mg of lemborexant once per day.
- the first dose is 20 mg, 22.5 mg, 25 mg, 27.5 mg or 30 mg of lemborexant once per day
- the second dose is 10 mg, 12.5 mg, 15 mg, or 17.5 mg of lemborexant once per day
- the third dose is 5 mg or 7.5 mg of lemborexant once per day.
- the method comprises administering lemborexant to the subject for at least 24 months, 30 months, or 36 months.
- Another aspect of the present disclosure relates to a method of selecting a subject having Alzheimer’s disease (AD) or at risk of developing AD for treatment with lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, comprising: (a) obtaining from the subject a measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglial response, and biomarker expression; (b) comparing the measurement from the subject to a measurement from a reference; and (c) selecting the subject for treatment with lemborexant if the measurement from the subject differs from the measurement from the reference.
- AD Alzheimer’s disease
- a pharmaceutically acceptable salt thereof or a solvate thereof
- the subject has mild cognitive impairment or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment.
- the subject is at risk for A ⁇ accumulation. In some embodiments, the subject is an ApoE4 carrier. In some embodiments, the subject has intermediate levels of amyloid PET (e.g., 20-40 centiloids). In some embodiments, the subject has elevated levels of amyloid PET (e.g., > 40 centiloids). [053] In some embodiments, the subject has early-stage AD. In some embodiments, the subject has pre-AD.
- the subject has been diagnosed with AD, based on brain imaging, cognitive function, and/or biomarker criteria.
- obtaining at least one measurement comprises obtaining data from a brain scan of the subject and/or obtaining data from a biological sample from the subject.
- the data from the brain scan indicates a level of tau phosphorylation, tau aggregation, A ⁇ plaque burden, and/or microglial response.
- the biological sample is a body fluid.
- the body fluid is cerebrospinal fluid (CSF), blood, or saliva.
- the reference is a control.
- the reference is a measurement from a control subject administered a placebo.
- the control does not have AD.
- the measurement from the subject is higher than the measurement from the control who does not have AD.
- the measurement from the subject is lower than the measurement from the control who does not have AD.
- the control has AD.
- the measurement from the subject is comparable to or higher than the measurement from the control who has AD.
- the measurement from the subject is comparable to or lower than the measurement from the control who has AD.
- the measurement of tau phosphorylation comprises a measurement of phosphorylation on one more of T181, T217, S202, S205, or T231.
- the measurement of tau aggregation comprises a measurement of insoluble tau aggregates (e.g., neurofibrillary tangles (NFTs)).
- the measurement of neurodegeneration comprises a measurement of cortical thickness and/or hippocampal volume or a measurement of loss of pyramidal neurons or granule neurons.
- the measurement of A ⁇ plaque burden comprises a measurement of A ⁇ plaque volume and/or growth of A ⁇ plaque volume.
- the measurement of A ⁇ plaque burden comprises a measurement of amyloid PET signal in a brain region of the subject or a measurement of A ⁇ in the CSF of the subject.
- the measurement of microglial response is a change in the expression of at least one microglial marker.
- the microglial marker is Iba1, Clec7a, CD68, TMEM119, or P2RY12.
- the measurement of microglial response is a measurement of phagocytosis by microglia.
- a further aspect of the present disclosure relates to a method of monitoring treatment efficacy in a subject having Alzheimer’s disease (AD) or at risk of developing AD, comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (b) administering to the subject a dose of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; and (d) comparing the second measurement from the subject to the first measurement from the subject, wherein a difference between the first measurement and the second measurement indicates effective treatment with lemborexant.
- AD Alzheimer’s disease
- Another aspect of the present disclosure relates to a method of treating a subject having Alzheimer’s disease (AD) or at risk of developing AD, comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglial response, and biomarker expression; (b) administering to the subject a first dose of Attorney Docket No.: 08061.0057-00304 lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (d) comparing the second measurement from the subject to the first measurement from the subject, and (e) administering a second dose of lemborexant if the first measurement differs from the second measurement.
- AD Alzheimer’s disease
- obtaining at least one measurement comprises obtaining data from a brain scan of the subject and/or obtaining data from a biological sample from the subject.
- the data from the brain scan indicates a level of tau phosphorylation, tau aggregation, A ⁇ plaque burden, and/or microglial response.
- the biological sample is a body fluid.
- the body fluid is cerebrospinal fluid (CSF), blood, or saliva.
- the first measurement from the subject is higher than the second measurement from the subject. In some embodiments, the first measurement from the subject is lower than the second measurement from the subject.
- the measurement of tau phosphorylation comprises a measurement of phosphorylation of one or more of T181, T217, S202, S205, or T231.
- the measurement of tau aggregation comprises a measurement of insoluble tau aggregates (e.g., neurofibrillary tangles (NFTs)).
- the measurement of neurodegeneration comprises a measurement of cortical thickness and/or hipoocampal volume or a measurement of loss of pyramidal neurons or granule neurons.
- the measurement of A ⁇ plaque burden comprises a measurement of A ⁇ plaque volume and/or growth of A ⁇ plaque volume.
- the measurement of A ⁇ plaque burden comprises a measurement of amyloid PET signal in a brain region of the subject or a measurement of A ⁇ in the CSF of the subject.
- the measurement of microglial response is a measure of expression of at least one microglial marker.
- the microglial marker is Iba1, Clec71, P2RY12 or TMEM 119.
- the measurement of microglial response is a measurement of phagocytosis by microglia.
- the measurement of a biomarker expression is a measurement of Ifnb1, MMP2, and/or Bace1 expression.
- the subject is amyloid-negative. In some embodiments, the subject has A ⁇ plaques. [071] In some embodiments, the subject has mild cognitive impairment and/or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment. [072] In some embodiments, the subject is at risk for further A ⁇ accumulation. In some embodiments, wherein the subject is an ApoE4 carrier. In some embodiments, wherein the subject has intermediate levels of amyloid PET (e.g., 20-40 centiloids).
- FIG.1 shows a study schema for a clinical trial.
- FIG.1 depicts an overview of the study design with a habitual bedtime of 22:00.
- FIGS.2A-H depicts data from the study of Example 2.
- FIG.2A shows a schematic illustration of the study design.
- FIG.2E shows representative spectograms of EEG and electromyography (EMG) analyses illustrating NREM, REM and wake patterns in vehicle-treated P301S/E4 and FIG.2F shows Lemborexant-treated P301S/E4 mice.
- White and dark bars note light phase and dark phase, respectively. Data represent mean ⁇ SEM; *p ⁇ 0.05, **p ⁇ 0.001, ***p ⁇ 0.0001.
- FIG.3 depicts data from the study of Example 2.
- FIG.3A shows representative images of AT8-stained phosphorylated tau at both serine 202 and threonine 205. Top panel show hippocampus and bottom show entorhinal and piriform cortices. Scale bar – 500 ⁇ m.
- FIG.3B shows representative images of MC1- stained tau. Scale bar – 500 ⁇ m.
- FIG.3G shows representative images of cresyl violet-stained brains used for volumetric analysis. Scale bar – 1mm.
- FIG.3H shows quantification of hippocampus volume and, FIG.3I shows piriform cortex volume.
- NfL plasma neurofilament light chain
- FIGS.4A-R depicts data from the study of Example 2.
- FIG.4A shows representative images of IBA1 (green), CD68 (red) and DAPI (blue) co-stained microglia of the CA3 region in hippocampus.
- FIG.4B shows representative images of TMEM119 (yellow) and DAPI (blue) stained microglia in hippocampal CA3 region.
- FIG.4C shows quantification of percentage IBA1 covered, and FIG.4D shows CD68 covered CA3.
- FIG.4E shows representative images of Clec7a (red) and DAPI (blue) stained microglia.
- FIG.4F shows quantification of percentage TMEM119 covered
- FIG.4 G shows Clec7a covered CA3.
- FIG.4H shows representative images of ApoE (green) and GFAP (red) costained astroglia.
- FIG.4I shows quantification of percentage ApoE colocalized with GFAP, or FIG.4J shows IBA1 in CA3.
- FIG.4K shows representative images of ApoE (green) costained with IBA1 (magenta) positive microglia.
- FIG.4L shows the percentage of Iba1-covered piriform/entorhinal cortex in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG. 4M shows the percentage of Iba1-covered dentate gyrus in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG.4N shows the percentage of Attorney Docket No.: 08061.0057-00304 TMEM119-covered piriform/entorhinal cortex in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG.4O shows the percentage of TMEM119-covered dentate gyrus in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG.4P shows the percentage of CD68-covered dentate gyrus in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG.4Q shows the percentage of CD68-covered piriform/entorhinal cortex in lemborexant-treated mice as compared to vehicle-treated control mice.
- FIG.4R shows a representative image of the CA1/CA2 region stained for GFAP, and
- FIG.4S shows plots of the percentage of GFAP-covered CA1/2
- FIG.4T shows plots of the dentate gyrus, and
- FIG.4U shows plots of the piriform/entorhinal cortex. Scale bar – 50 ⁇ m.
- FIG.5 depicts data from the study of Example 2.
- FIG 5A shows a volcano plot comparing differentially regulated genes in P301S/E4 mice treated with vehicle versus Lemborexant. Cut off value for significance is set at two-fold Log fold change.
- nE4 and nP301S/E4 10 mice/treatment group.
- FIG.5B shows a GO term analysis of genes significantly changed in P301S/E4 mice treated with vehicle versus Lemborexant.
- FIG.5C shows a heatmap illustrating all differentially expressed genes in P301S/E4 vehicle versus lemborexant that reached significance of Log10 adjusted p value ⁇ 0.05.
- FIGS.5D and 5E show representative images of VGLUT1 and PSD95-stained synapses in CA3. Scale bar – 50 ⁇ m.
- FIG.5F shows quantification of percentage VGLUT1 puncta in CA3, and
- FIG.5G shows results in piriform cortex.
- FIG.5H shows quantification of percentage PSD95 puncta in CA3, and
- FIG.5I shows results in piriform cortex.
- nE4 16-19 mice/treatment group;
- nP301S/E4 16- Attorney Docket No.: 08061.0057-00304 19 mice/treatment group.
- FIG.6 depicts data from the study of Example 2.
- FIG.7G the top panel illustrates representative images of hippocampus including granule cell layer, and the bottom panel show piriform cortex including pyramidal cell layer.
- FIG. 7H shows the volumetric analysis of cresyl violet-stained hippocampus
- FIG.7I shows the hemibrain subtracted by ventricle
- FIG.7J shows the pyramidal cell layer of the piriform cortex
- FIG.7K shows the hippocampal granule cell layer.
- FIG.8 depicts data from the study of Example 2.
- FIG.8A shows representative images of DAPI, IBA1 and P2RY12-costained microglia in the CA3.
- FIG.8B shows percentage P2RY12 covered CA3 quantified and
- FIG.8C shows results in the dentate gyrus.
- nE4 and nP301S/E4 16-19 mice/treatment group.
- FIG.8C shows the percentage of P2RY12-covered dentate gyrus.
- FIG.8D shows the percentage of P2RY12-covered piriform/entorhinal cortex.
- FIG.9 from the study in Example 3, shows changes in sleep in APP/PS1dE9 mice that were administered doxepin or lemborexant.
- FIG.9A is a schematic of the experimental design.
- FIG.9B, FIG.9C, and FIG.9D show the effects on total sleep, light phase sleep, and dark phase sleep in mice treated with doxepin, lemborexant (10 mg or 30 mg), or a vehicle.
- FIG.10 from the study in Example 3, shows the fibrillar amyloid plaque burden in APP/PS1dE9 mice that were administered doxepin or lemborexant.
- FIG.10A is a schematic for the timing of treatment of the mice.
- FIG.10B shows representative images of brain sections stained with X34, which labels fibrillar amyloid plaques.
- FIG.10C shows quantification of plaque burden (% area X34 staining) in different brain regions. Error bars indicate mean ⁇ SEM, each dot is a mouse. P values from 1-way ANOVA are shown.
- FIG.11 from the study of Example 3, shows the total amyloid plaque burden in APP/PS1dE9 mice treated with doxepin or lemborexant, as shown in FIG. 10A.
- FIG.11A shows representative brain sections stained for total amyloid plaques burden using the anti-A ⁇ antibody HJ3.4.
- FIG.11B is a quantification of plaque Attorney Docket No.: 08061.0057-00304 burden (% area HJ3.4 staining) in different brain regions.
- FIG.12 shows APP processing/cleavage in APP/PS1dE9 mice treated with doxepin or lemborexant.
- FIG.12A shows the results of a representative Western blot for full-length APP and APP C-terminal fragments (CTF- ⁇ and - ⁇ ). Beta-tubulin is shown as a loading control.
- FIG.12B shows the quantification of band intensity. Error bars indicate mean ⁇ SEM, each dot is a mouse. P values from 1-way ANOVA are shown.
- FIG.13 shows peri-plaque microglial clustering in APP/PS1dE9 mice treated with doxepin or lemborexant.
- FIG.13A shows representative images of samples stained for plaques (X34) and microglia (Iba1). The volume of microglia around each plaque was calculated from Z-stacks of confocal images using Imaris software.
- FIG.13B shows quantification of plaque volume (to indicate that similar sized plaques were quantified across conditions), and peri-plaque Iba1 volume. Error bars indicate mean ⁇ SEM, each dot is a mouse. P values from 1-way ANOVA are shown.
- FIG.14 shows peri-plaque microglial CD68 expression in APP/PS1dEP mice in APP/PS1dEP mice treated with doxepin or lemborexant.
- FIG.14A show representative images of samples stained for plaques (X34), microglia (Iba1), phagosomes (CD68).
- FIG.14B shows Iba1- colocalized CD68 quantified around each plaque using Imaris software. Error bars indicate mean ⁇ SEM, each dot is the average of 8-10 plaques from a single mouse. P values from 1-way ANOVA are shown.
- FIG.14C shows the Iba1 volume ( ⁇ m 3 ).
- FIG.14D shows co-localization of Iba1 and CD68, measured as a percentage of Iba1.
- FIG.14E shows the volume of co-localized Iba1 and CD68.
- FIG.15 shows the effects of lemborexant treatment on gene expression. Expression of transcripts encoding Ifnb1, Rab5a, and Mmp2 showed significant differences after lemborexant treatment. Data is displayed as fold change (relative to the mean for VEH). Error bars indicate mean ⁇ SEM, each dot is a single mouse. P values from 1-way ANOVA are shown.
- FIG.16 shows microglial amyloid plaque phagocytosis in APP/PS1dEP mice treated with lemborexant.
- FIG.16A shows a schematic of the experimental design.
- FIG.16B shows the flow cytometry gating strategy.
- FIG.16C shows methoxy-X04 (MX04) positivity in a CD45-low, CD11b+ population isolated as likely microglia.
- FIG.17 shows the amyloid plaque growth in mice with pre-existing plaques.
- FIG.17A is a schematic of experimental design.
- FIG.17B shows representative images of MX04, thiazine Red, and overlay images. P values are shown from a 1-way ANOVA.
- FIG.17C shows representative images of amyloid plaques labeled with X34, microglia labeled with IBA1, and microglial phagosome labeled with CD68.
- FIG.17D shows the percentage of plaque volume growth in VEH and lemborexant-treated mice. P values are from Mann- Whitney U test, due to non-Gaussian distribution of data.
- FIG.17E shows the quantification of colocalized IBA1-CD68, displayed as a percentage of total IBA1 (total microglial) area. P values are from a 1-way ANOVA.
- FIG.17F shows the percentage of plaque volume growth in mice treated with lemborexant, doxepin, or a vehicle control.
- FIG.17G shows the quantification of colocalized IBA1-CD68, displayed as a % of total IBA1 (total microglial) area. All graphs display mean ⁇ SEM, and each dot is one mouse.
- FIG.18 shows the effects of lemborexant or doxepin on rhythmic activity patterns in arrhythmic Bmal1 KO mice.
- FIG.18B shows a quantification of circadian locomotor behavior during different portions of the experiment (LD is indicated by the shaded area, DD+LEM is area with the shaded arrow, and DD is the rest of the recording). Data analyzed by 2-way ANOVA with Tukey post-hoc test. DETAILED DESCRIPTION I. Definitions [092] The following are definitions of terms used in the present application. [093] As used herein, the singular terms “a,” “an,” and “the” include the plural reference unless the context clearly indicates otherwise.
- a and/or B when used in conjunction with open-ended language such as “comprising” can refer, in some embodiments, to A only (optionally including elements other than B); in other embodiments, to B only (optionally including elements other than A); in yet other embodiments, to both A and B (optionally including other elements); etc.
- “at least one” means one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related Attorney Docket No.: 08061.0057-00304 or unrelated to those elements specifically identified.
- “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
- “15 mg to 30 mg” is intended to encompass, for example, 15.0 mg, 15.5 mg, 16.0 mg , 16.5 mg, 17.0 mg, 17.5 mg, 18.0 mg, 18.5 mg, 19.0 mg, 19.5 mg, 20.0 mg, 20.5 mg, 21.0 mg, 21.5 mg, 22.0 mg, 22.5 mg, 23.0 mg, 23.5 mg, 24.0 mg, 24.5 mg, 25.0 mg, 25.5 mg, 26.0 mg, 26.5 mg, 27.0 mg, 27.5 mg, 28.0 mg, 28.5 mg, 29.0 mg, 29.5 mg, 30.0 mg, 15 mg to 15.5 mg, 15 mg to 16 mg, 15 mg to 17.5 mg, 17.5 mg to 21 mg, 15 mg to 28 mg, and so forth.
- Amyloid refers to aggregates of proteins which form a fibrillar morphology. Amyloids are often formed from long, unbranched fibers that are characterized by an extended ⁇ -sheet secondary structure, which are approximately 7-13 nm in width and a few micrometers in length. Amyloids are usually extracellular, and found in vivo; in addition, the fibers bind the dye Congo Red and then show Attorney Docket No.: 08061.0057-00304 green birefringence when viewed between crossed polarizers.
- Amyloid-forming proteins have been identified and associated with serious diseases, including amyloid- ⁇ peptide (A ⁇ ) with Alzheimer’s disease (AD), islet amyloid polypeptide (IAPP) with diabetes type 2, and prion protein (PrP) with the spongiform encephalopathies.
- AD Alzheimer’s disease
- IAPP islet amyloid polypeptide
- PrP prion protein
- Amyloid ⁇ 1-42 (A ⁇ 42) refers to an amyloid beta monomer from amino acid 1 to 42 of the full-length protein (Table 5, SEQ ID NO:13).
- Amyloid ⁇ 1- 40 refers to an amyloid beta monomer from amino acid 1 to 40 of the full- length protein (Table 5, SEQ ID NO:14).
- Amyloid levels from amyloid PET can be reported using the Centiloid method in “centiloid” units (CL). (Klunk WE et al. The Centiloid Project: standardizing quantitative amyloid plaque estimation by PET. Alzheimer’s Dement. 2015; 11:1–15 e1–4).
- the Centiloid method measures a tracer on a scale of 0 CL to 100 CL, where 0 is deemed the anchor-point and represents the mean in young healthy controls and 100 CL represents the mean amyloid burden present in subjects with mild to moderate severity dementia due to AD.
- centiloid thresholds may vary, for example may be refined, based on new or additional scientific information. (See, e.g., http://www.gaain.org/centiloid-project.)
- An elevated level of amyloid can be set relative to a baseline threshold in a healthy control determined according to methods known to a person of ordinary skill in the art.
- whether a subject is “amyloid positive” or “amyloid negative” may be determined based on whether the subject has a positive amyloid load.
- a subject is determined to be amyloid-positive or Attorney Docket No.: 08061.0057-00304 amyloid-negative as indicated by longitudinal positron emission tomography (PET) assessment of an amyloid imaging agent uptake into the brain.
- PET longitudinal positron emission tomography
- a subject is “amyloid negative” if the florbetapir amyloid PET SUVr negativity is below 1.17.
- a subject is determined to be amyloid-positive or amyloid-negative by a CSF assessment of the presence of amyloid pathology using assessments of markers such as A ⁇ 1-42 (e.g., a soluble CSF biomarker analysis), alone or in combination with another method such as PET measurement of brain amyloid.
- Methods for measuring A ⁇ 38, A ⁇ 40, and A ⁇ 42 are known in the art, such as assays using LC MS/MS. Methods may include the PrecivityAD TM assay (see, e.g., Kirmess et al., J. Clinica Chimica Acta 519: 267-275 (2021)) and the Sysmex assay (https://www.eisai.com/news/2019/news201990.html) for measuring A ⁇ 42 and A ⁇ 40 in a blood or plasma sample or a CSF sample.
- PrecivityAD TM assay see, e.g., Kirmess et al., J. Clinica Chimica Acta 519: 267-275 (2021)
- Sysmex assay https://www.eisai.com/news/2019/news201990.html
- a qualitative visual read of PET scans may be used to determine amyloid positive and amyloid negative by categorizing subjects as having either “normal” or “abnormal” uptake on the basis of the PET image pattern. Readers will have been trained and certified to recognize brain PET images with abnormal or normal patterns of uptake, or the detection of amyloid is done through a semi-quantitative or quantitative approach.
- a threshold will be set for quantitatively determining from a biomarker (e.g., serum or CSF) and/or PET scan whether an A ⁇ brain load indicates a subject is amyloid-positive or negative.
- a subject is determined to be amyloid-positive or amyloid- negative by an MRI.
- the whole brain or at least one area of the brain (for example, cortical gray matter (i.e., cortex), lateral ventricles, frontal lobe, parietal lobe, temporal lobe, occipital lobe, cingulate cortex, amygdala, piriform Attorney Docket No.: 08061.0057-00304 cortex, entorhinal cortex, hippocampus, hippocampal CA3 (pyramidal neurons), and/or hippocampal dentate gyrus (granule cell neurons)) is/are analyzed by MRI.
- a subject is determined to be amyloid-positive or amyloid-negative by retinal amyloid accumulation.
- a subject is determined to be amyloid-positive or amyloid-negative by behavioral/cognitive phenotypes.
- tau protein or “tau” encompasses all tau isoforms, whether full-length, truncated, or post-translationally modified. In many animals, including but not limited to humans, non-human primates, rodents, fish, cattle, frogs, goats, and chicken, tau is encoded by the gene MAPT. In humans, there are six isoforms of tau that are generated by alternative splicing of exons 2, 3, and 10 of MAPT. These isoforms range in length from 352 to 441 amino acids.
- Exons 2 and 3 encode 29-amino acid inserts each in the N-terminus (called N), and full-length human tau isoforms may have both inserts (2N), one insert (1N), or no inserts (0N). All full-length human tau isoforms also have three repeats of the microtubule binding domain (called R). Inclusion of exon 10 at the C-terminus leads to inclusion of a fourth microtubule binding domain encoded by exon 10. Hence, full-length human tau isoforms may be comprised of four repeats (4R) of the microtubule binding domain (exon 10 included) or three repeats (3R) of the microtubule binding domain (exon 10 excluded). Human tau may or may not be post-translationally modified.
- tau may be phosphorylated, ubiquinated, glycosylated, and glycated.
- human tau encompasses the (2N, 3R), (2N, 4R), (1N, 3R), (1N, 4R), (0N, 3R), and (0N, 4R) isoforms, isoforms that are N- and/or C-terminally truncated species thereof, and all post-translationally modified isoforms.
- Alternative splicing of the gene encoding tau similarly occurs in Attorney Docket No.: 08061.0057-00304 other animals. In animals where the gene is not identified as MAPT, a homolog may be identified by methods well known in the art.
- Phosphorylation of specific amino acids results in phosphorylated tau (p-tau) isoforms. Phosphorylation may occur at different residues, such as T111, S113, T181, S199, S202, S208, T153, T175, T205, S214, T217, and T231.
- p-tau encompasses all phosphorylated tau (p-tau) isoforms, such as but not limited to p-tau181, p-tau217 and p-tau231.
- a disease associated with tau deposition in the brain may be referred to as a “tauopathy” or “tau pathology.”
- a clinical sign of a tauopathy may be aggregates of tau in the brain, including but not limited to neurofibrillary tangles.
- Other methods can be used to detect in a subject or measure tau phosphorylation at one or more amino acid residue and optionally total tau.
- tau can be purified from blood or cerebrospinal fluid (CSF) obtained from a subject.
- CSF cerebrospinal fluid
- Methods to measure phosphorylation of tau include high-resolution mass spectrometry. Suitable types of mass spectrometers are known in the art.
- quadrupole time-of-flight
- ion trap ion trap
- Orbitrap hybrid mass spectrometers that combine different types of mass analyzers into one architecture
- Other methods for measuring p-tau (phosphor-tau) and t- tau (total-tau) are known in the art, such as assays using Liquid Chromatography with tandem mass spectrometry (LC-MS-MS).
- LC-MS-MS Liquid Chromatography with tandem mass spectrometry
- the measurement of p-tau and t-tau can also be determined by positron emission tomography (PET) using radiotracers.
- PET positron emission tomography
- the whole brain or at least one area of the brain can be analyzed by PET.
- cortical gray matter i.e., cortex
- “relative to placebo” refers to a comparison of a biomarker (p-tau, A ⁇ , etc.) between the same biomarker of a subject being administered lemborexant and another subject being administered a placebo (a substance that has no therapeutic effect).
- “relative to baseline” refers to a comparison of a biomarker (p-tau, A ⁇ , etc.) between the same biomarker of a subject being administered lemborexant and the same subject prior to treatment with lemborexant.
- “maintenance” refers to a subject having or keeping the same level or about the same amount of a biomarker (p-tau, A ⁇ , etc.) in a subject’s sample (CSF, blood, etc.) between two time points (one before the administration of lemborexant and the other after administration of lemborexant).
- MMSE refers to the Mini-Mental State Examination, a cognitive instrument commonly used for screening purposes, but also often measured longitudinally in AD clinical trials having a 30 point scale with higher scores indicating less impairment and lower scores indicating more impairment.
- seven items measuring orientation to time and place, registration, recall, attention, language and drawing were assessed. (Folstein, M.F. et al., “Mini- mental state. A practical method for grading the cognitive state of subjects for the clinician.” J. Psychiatr.
- PSQI refers to the Pittsburgh Sleep Quality Index, a self-rated questionnaire which assesses sleep quality and disturbances over a 1- month time interval.
- Nineteen individual items generate seven “component” scores: subjective sleep quality, sleep latency, sleep duration, habitual sleep efficiency, Attorney Docket No.: 08061.0057-00304 sleep disturbances, use of sleeping medication, and daytime dysfunction.
- the sum of scores for these seven components yields one global score, ranging from 0 to 21, where lower scores denote a healthier sleep quality.
- the questionnaire consists of eight dichotomous (yes/no) items related to the clinical features of sleep apnea.
- the total score ranges from 0 to 8.
- Patients can be classified for obstructive sleep apnea (OSA) risk based on their respective scores.
- OSA obstructive sleep apnea
- the sensitivity of STOP-Bang score ⁇ 3 to detect moderate to severe OSA (apnea-hypopnea index [AHI] > 15) and severe OSA (AHI > 30) is 93% and 100%, respectively.
- PSG refers to polysomnography, a standard diagnostic test for OSA.
- PSG provides an assessment of OSA as the frequency of apneas and hypopneas per hour of sleep (the apnea-hypopnea index or AHI).
- the severity of OSA is classified as follows: (a) none/minimal: AHI ⁇ 5 per hour; (b) mild: AHI ⁇ 5, but ⁇ 15 per hour; (c) moderate: AHI ⁇ 15, but ⁇ 30 per hour; and (d) severe: AHI ⁇ 30 per hour.
- Subjects with “preclinical AD” or “pre-AD” as described herein, are cognitively normal (e.g., unimpaired) individuals with intermediate or elevated levels of amyloid in the brain. At least two clinical pre-AD states in which subjects have unimpaired cognition have been defined by IWG criteria: pre-symptomatic AD and asymptomatic AD (or “asymptomatic at risk”).
- Pre-symptomatic AD refers to subjects with an autosomal dominant monogenic mutation for AD, e.g., a mutation in Amyloid precursor protein (APP), Presenilin 1 (PSEN1), or Presenilin 2 (PSEN2), who are cognitively unimpaired. Subjects who are pre-symptomatic but carry the he autosomal dominant monogenic mutation will most likely develop AD.
- Asymptomatic refers to subjects who do not have clinical signs and symptoms of AD, but have the presence of one or more biomarkers of AD pathology. Stages of asymptomatic at risk may be further classified. Episodic memory and executive function deficits may emerge later. Accordingly, subjects with pre-AD may be identified by cognitively unimpaired asymptomatic stages.
- Cognitively normal can include individuals who are CDR 0, or individuals within the normal ranges of cognitive test scores (MMSE, International Shopping List Task, Logical Memory, etc.).
- Preclinical AD occurs prior to significant irreversible neurodegeneration and cognitive impairment and is typically characterized by the appearance of in vivo molecular biomarkers of AD and the absence clinical symptoms.
- Preclinical AD biomarkers that may suggest the future development of Alzheimer’s disease include, but are not limited to, intermediate or elevated levels of amyloid in the brain as measured by amyloid or tau positron emission tomography (PET) (e.g., a centiloid measure of about 20-40, e.g., a measure of about 20-32).
- PET amyloid or tau positron emission tomography
- Additional biomarkers may be used alone or in conjunction, including one or more of cerebrospinal fluid level of A ⁇ 1-42 and/or A ⁇ 1- 42/1 -40 ratio, cerebrospinal fluid level of total tau, cerebrospinal fluid level of Attorney Docket No.: 08061.0057-00304 neurogranin, cerebrospinal fluid level of neurofilament light peptide (NfL), and biomarkers measured in the serum or plasma (e.g.
- a ⁇ 1-42 the ratio of two forms of amyloid-b peptide (e.g., a ratio of A ⁇ 1-42/1-40 ratio of between about 0.092- 0.094 or below about 0.092), plasma levels of plasma total tau (T-tau), levels of phosphorylated tau (P-tau) isoforms (including tau phosphorylated at 181 (P-tau181), 217 (P-tau217), and 231 (P- tau231)), glial fibrillary acidic protein (GFAP), and neurofilament light (NfL)).
- T-tau plasma total tau
- P-tau levels of phosphorylated tau isoforms
- GFAP glial fibrillary acidic protein
- NfL neurofilament light
- certain risk factors contribute to development of AD.
- AD apolipoprotein E
- Other risk factors associated with AD include, but are not limited to, having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, suffering from other conditions such as obesity, diabetes, heart and/or blood vessel disease, cancer, and/or immune system dysfunction, and/or a sleep disorder such as insomnia or a circadian rhythm sleep disorder, or having lifestyle risk factors such as smoking, alcohol consumption, lack of exercise, lack of cognitive activity, and malnutrition, and having exposure to environmental risk factors such as air pollution, metals (e.g., aluminum, copper, and zinc).
- metals e.g., aluminum, copper, and zinc
- “Early AD” or “early Alzheimer’s disease,” (EAD) as used herein, is a continuum of AD severity from mild cognitive impairment due to AD - intermediate likelihood to mild Alzheimer’s disease dementia.
- Subjects with early AD include subjects with mild Alzheimer’s disease dementia as defined herein and subjects with mild cognitive impairment (MCI) due to AD - intermediate likelihood as defined Attorney Docket No.: 08061.0057-00304 herein.
- subjects with early AD have MMSE scores of 22 to 30 and Clinical Dementia Rating (CDR) global range 0.5 to 1.0.
- NIA-AA National Institute of Aging- Alzheimer’s Association
- McKhann, G.M. et al “The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging - Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease.” Alzheimer Dement. 2011; 7:263-9.
- a subject with early AD has evidence of elevated amyloid in the brain or a positive amyloid load.
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by PET assessment.
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by a CSF assessment of markers such as A ⁇ 1-42 (e.g., a soluble CSF biomarker analysis).
- markers such as A ⁇ 1-42
- subjects with AD may be selected according to the methods in WO 2023/283650, the contents of which are incorporated herein by reference.
- diagnostic thresholds may be identified by amyloid PET either by visual read (per the label of approved PET tracers) or by establishing a centiloid threshold above which subjects are considered to have elevated amyloid (e.g., varying between 15-40 centiloids).
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by measuring the concentration of A ⁇ 42 Attorney Docket No.: 08061.0057-00304 and a concentration of A ⁇ 40 and calculating a ratio of A ⁇ 42 to A ⁇ 40 (A ⁇ 42/40 ratio).
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by an MRI or PET.
- elevated amyloid in the brain or a positive amyloid load is indicated by retinal amyloid accumulation.
- more than one assessment method is used.
- Subjects with “mild Alzheimer's disease dementia,” as used herein, are subjects meet the NIA-AA core clinical criteria for probable Alzheimer's disease dementia in McKhann, G. M. et al., “The diagnosis of dementia due to Alzheimer's disease: Recommendations from the National Institute on Aging—Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease.” Alzheimer Dement.2011; 7:263-9.
- the subject has “elevated amyloid” or “intermediate amyloid.”
- amyloid levels from amyloid PET can be reported using the Centiloid method in “centiloid” units (CL).
- CL centiloid units
- the Centiloid method measures a tracer on a scale of 0 CL to 100 CL, where 0 is deemed the anchor-point and represents the mean in young healthy controls and 100 CL represents the mean amyloid burden present in subjects with mild to moderate severity dementia due to AD.
- centiloid thresholds may vary, for example may be refined, based on new or additional scientific information. (See, e.g., http://www.gaain.org/centiloid-project.)
- An elevated level of amyloid can be set relative to a baseline threshold in a healthy control determined according to methods known to a POSA.
- a centiloid Attorney Docket No.: 08061.0057-00304 value of 32.5 can be used as a threshold value for “elevated amyloid,” and an “intermediate amyloid” level refers to an A ⁇ amyloid PET in the range of 20-32.5 CL (e.g., 30 CL).
- a centiloid value of 40 can be used as a threshold value for “elevated amyloid,” and an “intermediate amyloid” level refers to an A ⁇ amyloid PET in the range of 20-40 CL.
- Subjects with “MCI due to AD - intermediate likelihood,” as used herein are those identified as such in accordance with the NIA-AA core clinical criteria for mild cognitive impairment due to Alzheimer's disease—intermediate likelihood. For example, symptomatic but not demented AD subjects with evidence of brain amyloid pathology making them less heterogeneous and more similar to mild Alzheimer's disease dementia subjects in cognitive and functional decline as measured by the ADCOMS Composite Clinical Score defined herein.
- ADAS-cog refers to Alzheimer's Disease Assessment Scale-Cognitive.
- the ADAS-cog is a widely used cognitive scale in Alzheimer's disease trials having a structured scale that evaluates memory (word recall, delayed word recall, and word recognition), reasoning (following commands), language (naming, comprehension), orientation, ideational praxis (placing letter in envelope) and constructional praxis (copying geometric designs).
- CDR-SB Clinical dementia rating—sum of boxes.
- the CDR is a clinical scale that describes 5 degrees of impairment in performance on each of 6 categories of function including memory, orientation, judgment and problem solving, community affairs, home and hobbies, and personal care.
- the ratings of degree of impairment obtained on each of the 6 categories of function are synthesized into 1 global rating of dementia CDR score (ranging from 0 to 3).
- a sum of boxes score provides an additional measure of change where each category has a maximum possible score of 3 points and the total score is a sum of the category scores giving a total possible score of 0 to 18 with higher scores indicating more impairment.
- the global score may be used as a clinical measure of severity of dementia.
- ADCOMS refers to Alzheimer's Disease Composite Score, a composite clinical score based on an analysis of four ADAS-Cog items (delayed word recall, orientation, word recognition, and word finding difficulty), two MMSE items (orientation to time, and drawing), and all six CDR-SB items (personal care, community affairs, home and hobbies, memory, orientation, and judgment and problem solving), as discussed in the Examples and in Wang, J. et al., “ADCOMS: a composite clinical outcome for prodromal Alzheimer's disease trials.” J. Neurol. Neurosurg. Psychiatry.2016; 87:993-999.
- ADCOMS was developed to be particularly sensitive to disease progression during early stages of AD, i.e., prodromal and mild AD.
- “ApoE4-positive” subjects and “ApoE4 carriers” refer to subjects who harbor the ⁇ 4 variant of the apolipoprotein gene.
- the ⁇ 4 variant is one of several major alleles of the apolipoprotein gene. The gene is generally responsible for metabolism of fats. It has been found that carriers of the apolipoprotein ⁇ 4 show significantly greater rates of amyloid retention when compared to non-carriers. (Drzezga, A.
- the subject is a heterozygous carrier of the apolipoprotein E ⁇ 4 gene allele. In some embodiments, the subject is a homozygous carrier of the apolipoprotein E ⁇ 4 gene allele.
- the term “clinical decline” refers to a worsening of one or more clinical symptoms of AD. Methods for measuring clinical decline may employ the tests and assays specified herein. In some embodiments, clinical decline is determined by a worsening of ADCOMS.
- clinical decline is determined by a worsening of MMSE. In some embodiments, clinical decline is determined by a worsening of ADAS-Cog. In some embodiments, clinical decline is determined by a worsening of FAQ. In some embodiments, clinical decline is determined by a worsening of CDR-SB. In some embodiments, clinical decline is determined by a worsening of Wechsler Memory Scale-IV Logical Memory (subscale) I and/or (subscale) II. In some embodiments, clinical decline is determined by a worsening of CDR score.
- clinical decline refers to a worsening in one or more biomarkers of AD or brain measurement (e.g., by PET or MRI), e.g., of brain atrophy and/or amyloid accumulation.
- the term “treat” also “treating” or “treatment” refers to any administration or application of a therapeutic agent to a subject having a Attorney Docket No.: 08061.0057-00304 disease or disorder, and includes inhibiting the disease, slowing progression of the disease, delaying progression, arresting its development, reversing progression of disease (e.g., reversing build up of A ⁇ fibrils), preventing the onset of the disease or at least one symptom of the disease, or preventing further development of the disease, relieving or ameliorating one or more symptoms or underlying condition(s) of the disease, curing the disease, improving one or more clinical metrics, or preventing reoccurrence of one or more symptoms of the disease.
- treating may comprise maintaining (i.e., preventing from worsening in) the severity of at least one symptom of the disease, for example, when the symptom would otherwise be expected to progress and/or worsen in the absence of administration or application of a therapeutic agent to a subject.
- maintaining the symptom may refer to the absence of a change (e.g., no significant change such as no statistically significant change) in the symptom after administration or application of the therapeutic agent to the subject, as compared to a control (e.g., a subject not receiving a therapy or receiving a placebo), for whom a change occurs (e.g., a significant change such as a statistically significant change) in the symptom.
- Complete treatment is not required.
- treatment of AD in a subject comprises an administration, e.g., an intravenous infusion, of a dual orexin receptor antagonist, e.g., lemborexant, e.g., to a subject at risk of developing AD but who does not yet show evidence of dementia.
- a dual orexin receptor antagonist e.g., lemborexant
- the composition may be administered to a subject at risk of developing Alzheimer's disease (e.g., based on a biomarker and/or family history), to a subject having one or more preclinical Attorney Docket No.: 08061.0057-00304 symptoms but not clinical symptoms of Alzheimer's disease, and/or to a subject reporting one or more of the physiological symptoms of Alzheimer's disease, even though a clinical diagnosis of having Alzheimer's has not been made.
- prevention may further include therapeutic benefit, by which is meant eradication or amelioration of the underlying condition being treated or of one or more of the physiological symptoms associated therewith. Prevention also encompasses arresting or slowing the further progression of one or more symptoms of a disease.
- control refers to a biological sample obtained from a subject that is distinct from those samples being evaluated and has a known AD status.
- the control samples are obtained from a subject who does not have a diagnosis of Alzheimer’s disease, e.g., according to one or more of the definitions above.
- the control samples may be obtained from a subject who does not have clinical symptoms of AD (e.g., cognitive impairment and/or dementia), and/or does not have any of the markers of AD pathology (e.g., PET scan or CSF analysis for biomarkers such as amyloid or tau).
- a control sample may be obtained from a healthy subject.
- a control may be obtained from a subject with co-morbidities that are not associated with AD. In some embodiments, the control may be obtained from a subject with a diagnosis along the spectrum of AD disease including, for example, preclinical AD or mild cognitive impairment. In some embodiments, a control sample may be a baseline sample collected from a subject prior to initiation of any treatment. In some embodiments, a control sample may be a sample collected from a control subject administered a placebo.
- a “therapeutically effective amount” refers to an amount of a compound or pharmaceutical composition sufficient to product a Attorney Docket No.: 08061.0057-00304 desired therapeutic effect, e.g., to reverse, arrest, delay, or slow a cognitive decline, and/or to reverse, arrest, delay, or slow the rate of change in one or more biomarkers of AD.
- a therapeutically effective amount of lemborexant administered to a subject may depend upon a number of factors including pharmacodynamic characteristics, route of administration, frequency of treatment, and health, age, and weight of the subject to be treated and, with the information disclosed herein, will be able to determine the appropriate amount for each subject. II.
- lemborexant affects at least one marker of AD pathology.
- lemborexant s surprising effects on AD and AD pathology may be related to lemborexant’s role in regulating sleep and treating insomnia, among other potential effects on AD pathology.
- lemborexant provides benefits not seen by other sleep agents such as doxepin, another drug approved for treating insomnia.
- doxepin another drug approved for treating insomnia.
- lemborexant and doxepin showed differential effects on A ⁇ plaque development, activation of phagocytic microglia, and expression of biomarkers Attorney Docket No.: 08061.0057-00304 involved in membrane receptor trafficking and inflammatory activity.
- lemborexant reduced total amyloid plaque burden (including both diffuse and fibrillar plaques), whereas doxepin reduced only total amyloid burden and not fibrillar plaque burden (Example 3, Section B).
- Lemborexant also increased activation of phagocytic microglia surrounding A ⁇ plaques, whereas doxepin did not (Example 3, Section E).
- lemborexant significantly upregulated expression of Ifnb1, an inflammatory mediator IFN-beta, the lysosomal protein Rab5a, and the A ⁇ degrading enzyme Mmp2, whereas doxepin did not (Example 3, section F).
- a method of reducing or maintaining the amount of p-tau and/or t-tau in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- the amount of p-tau and/or t-tau in the subject’s CSF is decreased or maintained after administration of a therapeutically effective amount of lemborexant as compared to the amount of p-tau and/or t-tau in the subject’s CSF prior to such administration.
- the amount of p-tau and/or t-tau in the subject’s CSF is decreased or maintained after administration of a therapeutically effective amount of lemborexant as compared to the amount of p-tau and/or t-tau in a subject’s CSF after administration of a placebo.
- Attorney Docket No.: 08061.0057-00304 [0133]
- Also disclosed herein is a method of reducing neurodegeneration in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- a method of reducing or maintaining amyloid beta in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- the subject in need of the inventive method(s) has demonstrated evidence of at least one disease chosen from Alzheimer’s disease, pre-Alzheimer’s disease, early Alzheimer’s disease, mild cognitive impairment, cerebral amyloid angiopathy, frontotemporal dementia, dementia with Lewy bodies, Lewy body dementia, Parkinson’s disease, vascular dementia, limbic- predominant age-related TDP-43 encephalopathy, frontotemporal lobar degeneration, corticobasal degeneration, Pick’s disease, multiple system atrophy, and progressive supranuclear palsy. III.
- the subject in need of one or more of the disclosed methods has demonstrated evidence of at least one disease chosen from Alzheimer’s disease, pre-Alzheimer’s disease, early Alzheimer’s disease, mild cognitive impairment, cerebral amyloid angiopathy, frontotemporal dementia, dementia with Lewy bodies, Lewy body dementia, Parkinson’s disease, vascular dementia, limbic-predominant age-related TDP-43 encephalopathy, frontotemporal lobar degeneration, corticobasal degeneration, Pick’s disease, multiple system atrophy, and progressive supranuclear palsy.
- Alzheimer’s disease pre-Alzheimer’s disease
- early Alzheimer’s disease mild cognitive impairment
- cerebral amyloid angiopathy dementia with Lewy bodies
- Lewy body dementia Lewy body dementia
- Parkinson’s disease vascular dementia
- frontotemporal lobar degeneration corticobasal degeneration
- Pick’s disease multiple system atrophy, and progressive supranuclear palsy.
- the subject in need thereof has demonstrated evidence of at least one disease chosen from Alzheimer’s disease, pre-Alzheimer’s disease, and early Alzheimer’s disease.
- the subject in need thereof has demonstrated evidence of mild Attorney Docket No.: 08061.0057-00304 cognitive impairment.
- the subject in need thereof has demonstrated evidence of cerebral amyloid angiopathy, frontotemporal dementia, dementia with Lewy bodies, Lewy body dementia, vascular dementia, limbic- predominant age-related TDP-43 encephalopathy, frontotemporal lobar degeneration, corticobasal degeneration.
- the subject in need thereof has demonstrated evidence of at least one disease chosen from Parkinson’s disease, Pick’s disease, multiple system atrophy, and progressive supranuclear palsy.
- One aspect of the present disclosure relates to a method for treating Alzheimer’s disease (AD) in a subject who has AD or is at risk for developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, thereby treating AD.
- the subject is in need of treatment (e.g. has AD, pre-AD, or is otherwise at risk of developing AD).
- treating AD refers one or more of inhibiting, slowing progression of, slowing a rate of progression of, delaying progression of, arresting development of, and reversing progression of AD, AD pathology, symptoms of AD, and/or underlying conditions of AD.
- treating refers to preventing the onset of or preventing development of AD, AD pathology, symptoms of AD, and/or underlying conditions of AD.
- treating refers to relieving or ameliorating one or more symptoms or underlying condition(s) of AD (e.g., ameliorating build-up of A ⁇ fibrils) and/or improving one or more clinical metrics of AD (e.g., cognitive function, brain amyloid or tau levels, and/or expression of a biomarker).
- treating refers to preventing occurrence or reoccurrence of one or more symptoms of AD.
- Attorney Docket No.: 08061.0057-00304 [0138]
- treating AD comprises reducing, halting, or slowing cognitive decline.
- a subject in need of treatment described herein is a subject who has AD, e.g., who has been diagnosed with AD. The diagnosis may be based on cognitive evaluation. The diagnosis may be based on measurements of AD pathology obtained by brain imaging (e.g., amyloid PET or tau PET) and/or expression of biomarkers in the subject.
- biomarkers comprise brain amyloid level, brain tau level, cerebrospinal fluid level of A ⁇ 1-42, cerebrospinal fluid level of total tau, cerebrospinal fluid level of neurogranin, and cerebrospinal fluid level of neurofilament light chain (NfL).
- the subject who has AD shows cognitive impairment and AD pathology. For example, a subject who has AD may have t-tau levels above 400 ng/L, A ⁇ 1-42 levels below 550 ng/L, and/or a A ⁇ 1-42/A ⁇ 1-40 ratio below 0.065.
- the subject in need of treatment has early Alzheimer’s disease (also called “Early AD” or “EAD”).
- a subject with early AD has symptoms on a continuum of AD severity ranging from mild cognitive impairment due to AD - intermediate likelihood to mild Alzheimer’s disease dementia. In some embodiments, a subject with early AD has mild Alzheimer’s disease dementia as defined herein and/or mild cognitive impairment (MCI) due to AD - intermediate likelihood as defined herein. In some embodiments, a subject has an MMSE score of 22 to 30 and Clinical Dementia Rating (CDR) global range 0.5 to 1.0. In some embodiments, a subject with early AD has evidence of elevated amyloid in the brain or a positive amyloid load. In some embodiments, elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by PET assessment.
- elevated amyloid in the brain or a Attorney Docket No.: 08061.0057-00304 positive amyloid load is indicated and/or confirmed by a CSF assessment of markers such as A ⁇ 1-42 (e.g., a soluble CSF biomarker analysis).
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by measuring a ratio of A ⁇ 42 to A ⁇ 40 (A ⁇ 42/40 ratio).
- elevated amyloid in the brain or a positive amyloid load is indicated and/or confirmed by an MRI.
- elevated amyloid in the brain or a positive amyloid load is indicated by retinal amyloid accumulation.
- the subject has pre-clinical Alzheimer’s disease (also called “pre-AD”).
- pre-AD may be cognitively normal, with intermediate or elevated levels of amyloid in the brain.
- a subject who has pre-AD may be identified by asymptomatic stages with or without memory complaints and emerging episodic memory and executive function deficits.
- the subject who has pre-AD has a CDR 0 and/or scores within the normal ranges of cognitive test scores (e.g., MMSE, International Shopping List Task, Logical Memory, etc.).
- the subject has other biomarkers that suggest the future development of AD, such as one or more of intermediate or elevated levels of amyloid in the brain by amyloid or tau positron emission tomography (PET) (e.g., a centiloid measure of about 20-40, e.g., a measure of about 20-32), cerebrospinal fluid level of A ⁇ 1-42 and/or A ⁇ 1-42/1 -40 ratio, cerebrospinal fluid level of total tau, cerebrospinal fluid level of neurogranin, cerebrospinal fluid level of neurofilament light peptide (NfL), and blood biomarkers as measured in the serum or plasma (e.g.
- PTT amyloid or tau positron emission tomography
- the ratio of two forms of amyloid-b peptide (A ⁇ 1-42/1-40 ratio, e.g., a ratio of between about 0.092-0.094 or below about 0.092), plasma levels of plasma total tau (T-tau), levels of Attorney Docket No.: 08061.0057-00304 phosphorylated tau (P-tau) isoforms (including tau phosphorylated at 181 (P-tau181), 217 (P-tau217), and 231 (P- tau231)), glial fibrillary acidic protein (GFAP), and neurofilament light (NfL)).
- the subject who has pre-AD may have intermediate amyloid (e.g., approximately 20-40 centiloids).
- the subject who has pre-AD may have elevate amyloid (e.g., > 40 centiloids).
- the subject may be at risk of developing AD.
- a subject may have one or more risk factors for developing AD, such as carrying a familial AD gene (e.g., the apolioprotein E ⁇ 4 allele, also called “APOE4” or “ApoE4”), having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, suffering from other conditions such as obesity, diabetes, heart and/or blood vessel disease, and more.
- a subject who has pre-AD is at risk of developing AD.
- the risk of developing AD may be a greater risk than a control subject for developing AD at all and/or for a greater risk for developing AD sooner than the time estimated for a control subject.
- a subject who has pre-AD and who is cognitively normal but has intermediate amyloid PET levels may be at risk for further A ⁇ accumulation and early spread of tau pathology over 4 years, as compared to a control subject.
- a subject who has pre-AD and who is cognitively normal but has elevated amyloid PET levels (>40 centiloids) may be at high risk for cognitive decline over 4 years, as compared to a control subject.
- treating AD comprises affecting a change (for example, slowing, delaying, or reducing) in at least one marker of AD pathology.
- the marker of AD pathology is a level of phosphorylation of tau, neurodegeneration, a change in microglial response, and/or presence of A ⁇ plaques.
- the marker of AD pathology may be present in a brain region in the subject, such as the hippocampus, somatomotor cortex, somatosensory cortex, piriform cortex, and/or entrorhinal cortex.
- the marker of AD pathology is detected in a brain scan.
- tau phosphorylation may be detected by tau PET; A ⁇ may be detected by amyloid PET.
- the marker of AD pathology is detected in a body fluid of the subject, such as blood (e.g., plasma) or CSF.
- various species of phosphorylated tau and A ⁇ may be detected in plasma or CSF from a subject.
- the subject does not show signs of dementia and/or cognitive impairment.
- the subject has mild cognitive impairment or mild dementia.
- the subject is amyloid positive.
- the subject may be at risk for further A ⁇ accumulation and/or spread of tau pathology.
- the subject may be at risk for cognitive decline.
- the subject may have intermediate levels of amyloid PET (e.g., approximately 20-40 centiloids). In some embodiments, the subject may have elevated levels of amyloid PET (e.g., > 40 centiloids). In some embodiments, the subject may carry the APOE4 gene. In some embodiments, the subject may have one or more risk factors for developing AD, such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease.
- risk factors for developing AD such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease.
- the ratio of the concentration of p-tau (also referred to herein as “phospho-tau” or “phosphorylated tau”) to the concentration of t-tau (also referred to herein as “total tau”) in the CSF of a subject can be used to assess the amount of phosphorylation of tau.
- the concentration of p-tau and of t-tau in the CSF of a subject is measured using liquid chromatography with tandem mass spectroscopy (LC MS/MS).
- the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is reduced compared to the ratio of CSF p-tau/t-tau of a subject to whom placebo was administered. In some embodiments, the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is maintained within (i.e., +/-) 10% of the ratio of CSF p-tau/t-tau of a subject to whom placebo was administered.
- the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is within 9%, within 8%, within 7%, within 6%, within 5%, within 4%, within 3%, within 2%, or within 1% of the ratio of CSF p-tau/t-tau of a subject to whom placebo was administered.
- the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is reduced compared to the ratio of CSF p-tau/t-tau of the subject prior to the administration of lemborexant.
- the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is maintained within (i.e., +/-) 10% of the ratio of CSF p-tau/t-tau of the subject prior to the administration of lemborexant.
- the ratio of CSF p-tau/t-tau in a subject to whom a therapeutically effective amount of lemborexant was administered is within 9%, within 8%, within 7%, within 6%, within 5%, within 4%, within 3%, within 2%, or within 1% of the ratio of CSF p-tau/t-tau of the subject prior to the administration of lemborexant.
- the concentration of amyloid beta (A ⁇ ) in the CSF is lower than the concentration of A ⁇ in the CSF of a subject to whom placebo was administered.
- the concentration of A ⁇ in the CSF is lower than the concentration of A ⁇ in the CSF of a subject prior to the administration of lemborexant. In some embodiments, the concentration of A ⁇ 38, A ⁇ 40, and/or A ⁇ 42 in the CSF is/are reduced. In some embodiments, the concentration of A ⁇ in the CSF is maintained in comparison to the concentration of A ⁇ in the CSF of a subject prior to the administration of lemborexant. In some embodiments, the concentration of A ⁇ 38, A ⁇ 40, and/or A ⁇ 42 in the CSF is/are maintained in comparison to the concentration of A ⁇ in the CSF of a subject prior to the administration of lemborexant.
- the amyloid PET signal in the brain of the subject to whom a therapeutically effective amount of lemborexant is administered is lower than the amyloid PET signal in the brain of a subject to whom Attorney Docket No.: 08061.0057-00304 placebo is administered.
- the amyloid PET signal in the brain of the subject to whom a therapeutically effective amount of lemborexant is administered is lower or maintained than the amyloid PET signal in the brain of a subject prior to administration of lemborexant.
- the increase in the concentration of A ⁇ in the CSF of a subject to whom lemborexant was administered is smaller than the increase in concentration of A ⁇ in the CSF of a subject to whom placebo was administered.
- the concentration of A ⁇ in the CSF of a subject to whom lemborexant was administered is at least 5% lower compared to a subject to whom placebo was administered. In some embodiments, the concentration of A ⁇ in the CSF of a subject to whom lemborexant was administered is at least 10%, at least 15%, at least 20%, or at least 25% lower compared to a subject to whom placebo was administered. [0155] In some embodiments, the concentration of A ⁇ in CSF is measured using liquid chromatography with tandem mass spectroscopy (LC MS/MS). In some embodiments, the concentration of A ⁇ 38, A ⁇ 40, and/or A ⁇ 42 in the CSF is measured using LC MS/MS.
- LC MS/MS liquid chromatography with tandem mass spectroscopy
- Methods for measuring A ⁇ 38, A ⁇ 40, and A ⁇ 42 are known in the art, such as assays using LC MS/MS. Methods may include the PrecivityAD TM assay (see, e.g., Kirmess et al., J. Clinica Chimica Acta 519: 267-275 (2021)) and the Sysmex assay (https://www.eisai.com/news/2019/news201990.html) for measuring A ⁇ 42 and A ⁇ 40 in a blood or plasma sample or a CSF sample.
- the concentration of A ⁇ in the CSF is measured using ELISA.
- the concentration of A ⁇ 38, A ⁇ 40, and/or A ⁇ 42 in the CSF is measured using ELISA.
- administration to a subject of a composition comprising a therapeutically effective amount of lemborexant results in a reduction of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, or at least 13% relative to baseline, of cerebrospinal fluid level concentration of A ⁇ 38, A ⁇ 40, and/or A ⁇ 42.
- p-tau and t-tau are reduced.
- p-tau, t-tau, and/or aggregated tau is reduced.
- the reduction of tau phosphorylation occurs in the whole brain or areas of the brain such as, for example, the frontal lobe, parietal lobe, temporal lobe, occipital lobe, cingulate cortex, amygdala, hippocampus, entorhinal cortex, and/or piriform cortex.
- the tau PET signal in the brain is decreased compared to placebo. In some embodiments, the tau PET signal in the brain is decreased or maintained compared to baseline.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of concentration of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject to whom placebo was administered. In some embodiments, administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of concentration of p-tau in the CSF of the subject compared to the concentration of p- tau in the CSF of the subject prior to the administration of lemborexant.
- administration to a subject of a therapeutically effective amount of lemborexant results in the maintenance of the amount of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject to whom Attorney Docket No.: 08061.0057-00304 placebo was administered.
- administration to a subject of a therapeutically effective amount of lemborexant results in the maintenance of the amount of p-tau in the CSF of the subject prior to the administration of lemborexant.
- the concentration of p-tau in the CSF is measured using liquid chromatography with tandem mass spectroscopy (LC MS/MS).
- administration to a subject of a therapeutically effective amount of lemborexant results in the maintenance of the amount of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject to whom placebo was administered.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, or at least 13% of the amount of p-tau in the CSF of the subject compared to the amount of p-tau in the CSF of the subject to whom placebo was administered.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, or at least 13% of the amount of p-tau in the CSF of the subject compared to the amount of p-tau in the CSF of the subject prior to the administration of lemborexant.
- the increase in the amount of p-tau in the CSF of a subject to whom lemborexant was administered is lower than the increase in amount of p-tau in the CSF of a subject to whom placebo was administered.
- Attorney Docket No.: 08061.0057-00304 [0161]
- administration to a subject of a therapeutically effective amount of lemborexant results in a maintenance of or reduction of concentration of p-tau in the CSF of the subject compared to the concentration of p- tau in the CSF of the subject at baseline through 18 months after administration of lemborexant.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, or at least 13% concentration of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject at baseline.
- the reduction of the amount of p-tau or t-tau in a subject’s CSF is due to a reduction or maintenance in the amount of p-tau or t-tau in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- the reduction of the amount of p-tau in a subject’s CSF is due to an increase in the dephosphorylation of p-tau.
- the reduction of the amount of p-tau in a subject’s CSF is due to a reduction of the amount of tau.
- the reduction of the amount of p-tau in a subject’s CSF is due to a reduction of ratio of p-tau/t-tau.
- administration to a subject of a therapeutically effective amount of lemborexant disclosed results in a reduction or maintenance of the concentration of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject to whom placebo was administered.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least about 5 pg/mL, at least about 10 pg/mL, at least about 15 pg/mL, at least about 20 pg/mL, at least about 25 pg/mL, at least Attorney Docket No.: 08061.0057-00304 about 30 pg/mL, at least about 35 pg/mL, or at least about 40 pg/mL, relative to placebo, of cerebrospinal fluid amount of p-tau.
- administration to a subject of a composition comprising a therapeutically effective amount of lemborexant disclosed herein results in a reduction of at least about 40 pg/mL, relative to placebo, of cerebrospinal fluid amount of p-tau.
- administration to a subject of a therapeutically effective amount of lemborexant disclosed results in a reduction or maintenance of the concentration of p-tau in the CSF of the subject compared to the concentration of p-tau in the CSF of the subject prior to the administration of lemborexant.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least about 5 pg/mL, at least about 10 pg/mL, at least about 15 pg/mL, at least about 20 pg/mL, at least about 25 pg/mL, at least about 30 pg/mL, at least about 35 pg/mL, or at least about 40 pg/mL, relative to baseline, of cerebrospinal fluid amount of p-tau.
- administration to a subject of a composition comprising a therapeutically effective amount of lemborexant disclosed herein results in a reduction of at least about 40 pg/mL, relative to baseline, of cerebrospinal fluid amount of p-tau.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least about 5 pg/mL, at least about 10 pg/mL, at least about 15 pg/mL, at least about 20 pg/mL, at least about 25 pg/mL, at least about 30 pg/mL, at least about 35 pg/mL, or at least about 40 pg/mL, relative to baseline, of cerebrospinal fluid amount of p-tau through 18 months after administration of the composition comprising a therapeutically effective amount of lemborexant.
- administration to a subject of a composition comprising a therapeutically effective amount of lemborexant results in Attorney Docket No.: 08061.0057-00304 a reduction of at least 40 pg/mL, relative to baseline, of cerebrospinal fluid amount of p-tau through 18 months after administration of the composition comprising a therapeutically effective amount of at least lemborexant.
- administration to a subject of a therapeutically effective amount of lemborexant results in a reduction of at least about 5 pg/mL, at least about 10 pg/mL, at least about 15 pg/mL, at least about 20 pg/mL, at least about 25 pg/mL, at least about 30 pg/mL, at least about 35 pg/mL, or at least about 40 pg/mL, relative to placebo, of cerebrospinal fluid amount of p-tau through 18 months after administration of the composition comprising a therapeutically effective amount of lemborexant.
- administration to a subject of a composition comprising a therapeutically effective amount of lemborexant results in a reduction of at least 40 pg/mL, relative to placebo, of cerebrospinal fluid amount of p-tau through 18 months after administration of the composition comprising a therapeutically effective amount of at least lemborexant.
- the amount of p-tau is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, the amount of p-tau is reduced within 24 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline.
- the amount of p-tau is reduced within 12 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, the amount of p-tau is reduced within 6 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. [0168] In some embodiments, the amount of t-tau is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to the Attorney Docket No.: 08061.0057-00304 subject’s baseline. In some embodiments, the amount of t-tau is reduced within 24 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline.
- the amount of t-tau is reduced within 12 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, the amount of t-tau is reduced within 6 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. [0169] In some embodiments, the amount of A ⁇ is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, the amount of A ⁇ is reduced within 24 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline.
- the amount of A ⁇ is reduced within 12 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, the amount of A ⁇ is reduced within 6 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. [0170] In some embodiments, the amount of p-tau in a subject’s CSF is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to a subject to whom placebo was administered. In some embodiments, the amount of tau phosphorylation in a subject’s CSF is reduced within 24 hours of administration of the first dose of lemborexant to the subject relative to a subject to whom placebo was administered.
- the amount of tau phosphorylation in a subject’s CSF is reduced within 12 hours of administration of the first dose of lemborexant to the subject relative to a subject to whom placebo was administered. In some embodiments, the amount of tau Attorney Docket No.: 08061.0057-00304 phosphorylation in a subject’s CSF is reduced within 6 hours of administration of the first dose of lemborexant to the subject relative to a subject to whom placebo was administered. A.
- a further aspect of the present disclosure relates to a method of altering tau (for example, reducing or delaying tau accumulation, tau phosphorylation, and/or tau spreading, and/or slowing a rate thereof) in a subject having AD or at risk for developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, wherein the therapeutically effective amount is sufficient to reduce alter tau in the subject.
- altering tau comprises reducing or delaying tau accumulation, tau phosphorylation, or tau spreading, or slowing a rate of any of these.
- altering tau is slowing progression of, slowing a rate of progression of, delaying progression of, arresting development of, and reversing progression of tau pathology (e.g, tau accumulation, tau phosphorylation, and/or tau spreading), for example, in a brain region.
- the brain region may be the cortex or the hippocampus.
- the brain region may be the CA1 region, CA2 region, CA3 region, and/or dentate gyrus of the hippocampus.
- the brain region may be the entorhinal cortex and/or the piriform cortex.
- altering tau is preventing the onset of or preventing development of tau pathology.
- Altering tau may reduce, delay, or slow a rate of onset and/or progression of symptoms of tau pathology.
- altering tau is relieving or ameliorating one or more symptoms of tau pathology and/or improving one or more clinical metrics of tau pathology (e.g., cognitive function, brain amyloid or tau levels, and/or expression of a Attorney Docket No.: 08061.0057-00304 biomarker).
- altering tau is preventing occurrence or reoccurrence of one or more symptoms of tau pathology.
- the subject is amyloid negative.
- the subject may have mild cognitive impairment or mild dementia.
- the subject does not show signs of dementia and/or cognitive impairment.
- the tau is altered relative to a reference. Accordingly, the methods described herein may comprise reducing and/or delaying tau accumulation, tau phosphorylation, and/or tau spreading, and/or slowing the rate of any of these, as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment. In some embodiments, the reference is a baseline measurement from a control subject. The reference may be a measurement obtained from more than one control subjects, which is used as a standard or threshold measurement. In some embodiments, the reference is a measurement from a control subject administered a placebo.
- the methods herein comprise altering (e.g., reducing, arresting, or slowing the growth of) tau in a brain region of the subject.
- Altering tau in the brain region may comprise altering a tau PET signal in the brain region.
- the brain region is the hippocampus, entorhinal cortex, and/or the piriform cortex.
- altering tau comprises altering tau in a body fluid of the subject.
- tau levels in the subject’s brain may be detected in the subject’s body fluid.
- the body fluid is blood (e.g., plasma) or CSF.
- one or more forms of tau may be altered.
- tau is total tau. In some embodiments, tau is aggregated tau. In Attorney Docket No.: 08061.0057-00304 some embodiments, the tau is a phosphorylated form of tau (phospho-tau). Phospho-tau may be tau that is phosphorylated on one or more of T181, T217, S202, S205, or T231. [0178] In some embodiments, altering tau comprises altering a ratio of phopho-tau to total tau. In some embodiments, the ratio of phospho-tau to total tau is altered such that the ratio is maintained within 10% of the ratio of phospho-tau to total tau of the subject prior to the administration of lemborexant.
- a rate of dephosphorylation of phospho-tau is increased. In some embodiments, a rate of phosphorylation of tau is decreased. In some embodiments, altering tau comprises altering tau within 48 hours of administration of a first dose of lemborexant. For example, tau may be reduced within 48 hours of administration of the first dose of lemborexant. In some embodiments, reducing tau comprises altering phospho-tau in the hippocampus, entorhinal cortex, and/or piriform cortex.
- a further aspect of the present disclosure relates to a method of maintaining tau (e.g., tau accumulation, tau phosphorylation, and/or tau spreading) in a subject having AD or at risk of developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, wherein the therapeutically effective amount is sufficient to maintain tau in the subject.
- maintaining tau refers to maintainingtau accumulation, tau phosphorylation, and/or tau spreading, when tau would be expected to progress and/or worsen in the absence of administration or application of a therapeutic agent to a subject.
- maintaining tau may refer to the absence of a change (e.g., no significant change such as no statistically significant change) in tau (e.g., tau accumulation, tau phosphorylation, and/or tau Attorney Docket No.: 08061.0057-00304 spreading) after administration or application of the therapeutic agent to the subject, as compared to a control, for whom administration or application of the therapeutic agent leads to a change (e.g., a significant change such as a statistically significant change) in tau.
- maintaining tau comprises maintaining tau pathology (e.g., tau accumulation, tau phosphorylation, and/or tau spreading), for example, in a brain region.
- the brain region may be the cortex or the hippocampus.
- the brain region may be the CA1 region, CA2 region, CA3 region, and/or dentate gyrus of the hippocampus.
- the brain region may be the entorhinal cortex and/or the piriform cortex.
- maintaining tau is preventing the onset of or preventing development of tau pathology, for example, because tau pathology is maintained without change. Maintaining tau may reduce, delay, or slow a rate of onset and/or progression of symptoms of tau pathology, for example, because tau pathology is maintained without change.
- maintaining tau may result in relieving or ameliorating one or more symptoms of tau pathology and/or improving one or more clinical metrics of tau pathology (e.g., cognitive function, brain amyloid or tau levels, and/or expression of a biomarker). In some embodiments, maintaining tau may result in preventing occurrence or reoccurrence of one or more symptoms of tau pathology.
- the subject is amyloid negative. The subject may have mild cognitive impairment or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment. [0183] In some embodiments, the subject shows signs of cognitive impairment. In some embodiments, the subject is amyloid positive. In some embodiments, the subject has a diagnosis of AD, e.g., early AD.
- the tau is maintained relative to a reference. Accordingly, the methods described herein may comprise maintaining tau accumulation, tau phosphorylation, and/or tau spreading (or the rate of any of these) as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference may be a measurement obtained from more than one control subject, and may be used as a standard or threshold measurement.
- the reference is a measurement from a control subject administered a placebo.
- maintaining tau comprises maintaining tau in a brain region of the subject.
- Maintaining tau in the brain region may comprise altering, reducing, or maintaining a tau PET signal in the brain region.
- the brain region is the hippocampus, entorhinal cortex, and/or the piriform cortex.
- maintaining tau comprises maintaining tau in a body fluid of the subject. Tau levels in the subject’s brain may be correlated with a level in the subject’s body fluid.
- the body fluid is blood (e.g., plasma) or CSF.
- one or more forms of tau may be maintained.
- tau is total tau.
- tau is aggregated tau.
- the tau is a phosphorylated form of tau (phospho-tau).
- Phospho-tau may be tau that is phosphorylated on one or more of T181, T217, S202, S205, or T231.
- maintaining tau comprises maintaining a ratio of phopho-tau to total tau.
- the ratio of phospho-tau to total tau is maintained within 10% of the ratio of phospho-tau to total tau of the subject prior to Attorney Docket No.: 08061.0057-00304 the administration of lemborexant.
- maintaining tau comprises maintaining tau within 48 hours of administration of a first dose of lemborexant.
- phospho-tau is maintained in the hippocampus, entorhinal cortex, and/or piriform cortex. V.
- administration to a subject of a therapeutically effective amount of lemborexant results in an increase in the number of activated microglial cells compared to placebo. In some embodiments, the increase in the number of activated microglial cells is measured by PET. In some embodiments, the activated microglial cells are phagocytic microglial cells. [0190] In some embodiments, administration to a subject of a therapeutically effective amount of lemborexant results in an increase in the number of activated microglial cells compared to baseline. In some embodiments, the increase in the number of activated microglial cells is measured by PET. In some embodiments, the activated microglial cells are phagocytic microglial cells.
- the whole brain or at least one area of the brain for example, cortical gray matter (i.e., cortex), lateral ventricles, frontal lobe, parietal lobe, temporal lobe, occipital lobe, cingulate cortex, amygdala, piriform cortex, entorhinal cortex, hippocampus, hippocampal CA3 (pyramidal neurons), and/or hippocampal dentate gyrus (granule cell neurons) is/are analyzed by PET.
- cortical gray matter i.e., cortex
- lateral ventricles for example, cortical gray matter (i.e., cortex), lateral ventricles, frontal lobe, parietal lobe, temporal lobe, occipital lobe, cingulate cortex, amygdala, piriform cortex, entorhinal cortex, hippocampus, hippocampal CA3 (pyramidal neurons), and/or hip
- One aspect of the present disclosure relates to a method of modulating a microglial response in a subject having Alzheimer’s disease (AD) or at risk for developing AD, comprising administering to the subject a therapeutically Attorney Docket No.: 08061.0057-00304 effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, wherein the therapeutically effective amount is sufficient to modulate the microglial response in the subject.
- modulating refers to increasing or decreasing a microglial response, and/or to increasing or decreasing a rate of a microglial response.
- the number of microglia do not change, but the response (e.g., activation, reactivation, reactivity, differentiation, etc.) of the microglia is modulated.
- the modulation may differ depending on brain region (e.g., microglial activation or other response may occur differently in different brain regions).
- modulation of the microglial response is measured in a subject and compared to the microglial response in a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference may be a measurement obtained from more than one control subjects, which is used as a standard or threshold measurement.
- the reference is a measurement from a control subject administered a placebo.
- modulating the microglial response comprises modulating expression of at least one microglial marker.
- the microglial marker may be a general microglial marker.
- the general microglial marker may be a general marker for microglia in a specific context or disease setting (e.g., a general marker for activated microglia, reactive microglia, and/or microglia in a disease setting).
- the general microglial marker may be Iba1, Clec7a, or CD68.
- the microglial marker may be a homeostatic microglial marker.
- the homeostatic microglial marker is TMEM119 or P2RY12.
- modulating the microglial response comprises modulating activity of phagocytic microglia.
- the subject has mild cognitive impairment or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment.
- the subject is amyloid-negative.
- the subject may have tau pathology.
- the subject may have neurodegeneration in a brain region, for example, the hippocampus, the entorhinal cortex, and/or the piriform cortex.
- the brain region is the CA1 region, the CA2 region, the CA3 region, or the dentate gyrus in the hippocampus.
- modulating the microglial response comprises modulating a response in microglia associated with degenerating neurons.
- Microglia associated with neurodegenerating neurons may be close in proximity to the neurons, for example, when observed in a scan or a sample obtained from the subject.
- microglia associated with degenerating neurons may phagocytose the degenerating neurons and/or debris therefrom.
- administering to these subjects a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof may comprise reducing expression of at least one general microglial marker.
- the general microglial marker may be Iba1, CD68, or Clec7a.
- modulating the microglial response comprises increasing expression of at least one homeostatic microglial marker.
- the homeostatic microglial marker may be TMEM119 or P2RY12.
- the subject is amyloid positive, e.g., the subject has A ⁇ plaques.
- the A ⁇ plaques may be fibrillar A ⁇ plaques.
- the A ⁇ plaques are present in the hippocampus, the somatomotor cortex, the somatosensory cortex, and/or the piriform cortex of the subject.
- the subject does not show signs of dementia and/or cognitive impairment.
- the subject has mild cognitive impairment or mild dementia.
- the subject is at risk for further A ⁇ accumulation.
- the subject may be at risk for spread of tau pathology.
- the subject may be at risk for cognitive decline.
- the subject may have intermediate levels of amyloid PET (e.g., approximately 20-40 centiloids).
- the subject may have elevated levels of amyloid PET (e.g., > 40 centiloids).
- the subject may be an ApoE4 carrier.
- the subject may have one or more risk factors for developing AD, such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease.
- the subject has early-stage AD.
- the subject has pre-AD.
- modulating the microglial response comprises modulating a response in microglia associated with A ⁇ plaques. Microglia associated with A ⁇ plaques may be close in proximity to the A ⁇ plaques, for example, when observed in a scan or a sample obtained from the subject.
- microglia associated with A ⁇ plaques may phagocytose the A ⁇ plaques.
- administering to these subjects a therapeutically effective amount of lemborexant, a Attorney Docket No.: 08061.0057-00304 pharmaceutically acceptable salt thereof, or a solvate thereof may comprise increasing expression of a general microglial marker.
- the general microglial marker may be Iba1, Clec7a, or CD68.
- modulating the microglial response comprises increasing phagocytosis of A ⁇ plaques by phagocytic microglia.
- modulating the microglial response comprises reducing expression of a homeostatic microglial marker.
- the homeostatic microglial marker may be TMEM119 or P2RY12.
- the homeostatic microglial marker may be TMEM119 or P2RY12.
- administration to a subject of a therapeutically effective amount of lemborexant results in a decrease or maintenance in amyloid plaques compared to placebo. In some embodiments, administration to a subject of a therapeutically effective amount of lemborexant results in a decrease or maintenance in fibrillar amyloid plaques compared to placebo. In some embodiments, administration to a subject of a therapeutically effective amount of lemborexant results in a decrease or maintenance in amyloid plaques compared to baseline.
- administration to a subject of a therapeutically effective amount of lemborexant results in a decrease or maintenance in fibrillar amyloid plaques compared to baseline.
- the amyloid plaques are fibrillar amyloid plaques.
- Altering A ⁇ plaques Another aspect of the present disclosure relates to a method of altering A ⁇ plaques (e.g., reducing or delaying formation of A ⁇ plaques, or slowing a rate of growth thereof) in a subject having AD or at risk for developing AD, comprising administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, Attorney Docket No.: 08061.0057-00304 wherein the therapeutically effective amount is sufficient to alter A ⁇ plaques in the subject.
- a therapeutically effective amount is sufficient to alter A ⁇ plaques in the subject.
- altering A ⁇ plaques comprises slowing progression of, slowing a rate of progression of, delaying progression of, arresting development of, and reversing progression of A ⁇ plaque formation and/or A ⁇ plaque growth.
- altering A ⁇ plaques comprises preventing the onset of or preventing development of A ⁇ plaque pathology (e.g., any pathology resulting from or coinciding with A ⁇ plaque formation and/or A ⁇ plaque growth). Altering A ⁇ plaques may reduce, delay, or slow a rate of onset and/or progression of symptoms of this pathology.
- altering A ⁇ plaques comprises relieving or ameliorating one or more symptoms of A ⁇ plaque pathology and/or improving one or more clinical metrics of A ⁇ plaque pathology (e.g., cognitive function, brain amyloid or tau levels, and/or expression of a biomarker).
- altering A ⁇ plaques comprises preventing occurrence or reoccurrence of one or more symptoms of A ⁇ plaque formation and/or A ⁇ plaque growth.
- the A ⁇ plaques are altered relative to a reference. Accordingly, altering A ⁇ plaques may comprise reducing and/or delaying A ⁇ plaque formation, and/or slowing the rate thereof, as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment.
- the reference is a baseline measurement from a control subject.
- the reference may be a measurement obtained from more than one control subjects, which is used as a standard or threshold measurement.
- the reference is a measurement from a control subject administered a placebo.
- Attorney Docket No.: 08061.0057-00304 [0209]
- the A ⁇ plaques are fibrillar plaques.
- the A ⁇ plaques are total plaques, and may comprise non-fibrillar (e.g., diffuse) plaques.
- altering A ⁇ plaques comprises reducing the growth or rate of growth of A ⁇ plaques.
- altering A ⁇ plaques comprises altering an amyloid PET signal obtained from a brain region of the subject.
- altering A ⁇ plaques corresponds to a reduction in the concentration of A ⁇ in the subject’s CSF.
- the A ⁇ may be A ⁇ 38, A ⁇ 40, and/or A ⁇ 42.
- altering A ⁇ comprises altering A ⁇ plaques within 48 hours of administration of a first dose of lemborexant.
- the subject does not show signs of dementia and/or cognitive impairment.
- the subject has mild cognitive impairment or mild dementia. [0213] In some embodiments, the subject is at risk for further A ⁇ accumulation. The subject may also be at risk for spread of tau pathology. The subject may be at risk for cognitive decline. In some embodiments, the subject may have intermediate levels of amyloid PET (e.g., approximately 20-40 centiloids). In some embodiments, the subject may have elevated levels of amyloid PET (e.g., > 40 centiloids). In some embodiments, the subject may be an ApoE4 carrier.
- the subject may have one or more risk factors for developing AD, such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from Attorney Docket No.: 08061.0057-00304 traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease.
- the subject has early-stage AD.
- the subject has pre-AD.
- Neurodegeneration Also disclosed herein is a method of reducing neurodegeneration in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- the subject in need thereof has demonstrated evidence of at least one disease chosen from Alzheimer’s disease, pre-Alzheimer’s disease, early Alzheimer’s disease, mild cognitive impairment, cerebral amyloid angiopathy, frontotemporal dementia, dementia with Lewy bodies, Lewy body dementia, Parkinson’s disease, vascular dementia, limbic- predominant age-related TDP-43 encephalopathy, frontotemporal lobar degeneration, corticobasal degeneration, Pick’s disease, multiple system atrophy, and progressive supranuclear palsy.
- the subject in need thereof has demonstrated evidence of at least one disease chosen from Alzheimer’s disease, pre-Alzheimer’s disease, and early Alzheimer’s disease.
- the subject in need thereof has demonstrated evidence of mild cognitive impairment.
- the subject in need thereof has demonstrated evidence of cerebral amyloid angiopathy, frontotemporal dementia, dementia with Lewy bodies, Lewy body dementia, vascular dementia, limbic- predominant age-related TDP-43 encephalopathy, frontotemporal lobar degeneration, corticobasal degeneration.
- the subject in need thereof has demonstrated evidence of at least one disease chosen from Parkinson’s Attorney Docket No.: 08061.0057-00304 disease, Pick’s disease, multiple system atrophy, and progressive supranuclear palsy.
- the reduction of neurodegeneration is observed by maintenance or slowing of reduction of the thickness of the cortex relative to a subject to whom placebo was administered. In some embodiments, the reduction of neurodegeneration is observed by maintenance or slowing of reduction of the thickness of the cortex relative to a subject’s baseline. In some embodiments, the reduction of neurodegeneration is observed by maintenance or slowing of reduction of the size of the hippocampus. In some embodiments, the reduction of neurodegeneration is observed by the preservation or reduction of loss of pyramidal neuronal cells relative to a subject’s baseline or to a subject to whom placebo was administered.
- the reduction of neurodegeneration is observed by the preservation or reduction of loss of granule neuronal cells relative to a subject’s baseline or to a subject to whom placebo was administered.
- Methods for measuring the thickness of portions of the brain such as the cortex or measuring the size of the hippocampus can be achieved using magnetic resonance imaging (MRI).
- MRI magnetic resonance imaging
- High spatial resolution sMRI now allows for volumetry of hippocampal subfields.
- Early changes in CA1 have been observed in AD, with volumetric studies indicating that CA1 atrophy measures may improve diagnostic accuracy at the MCI stage.
- Novel MRI techniques such as quantitative susceptibility mapping (QSM) or the T2* transverse relaxation time, have shown that iron levels and its rate of accumulation are heterogeneous in the human brain and correlates with cognitive impairment and slowing of motor performance.
- QSM quantitative susceptibility mapping
- T2* transverse relaxation time have shown that iron levels and its rate of accumulation are heterogeneous in the human brain and correlates with cognitive impairment and slowing of motor performance.
- Neuronal dysfunction and altered connectivity of distinct brain networks are thought to occur early in the course of neurodegenerative diseases and can be measured indirectly Attorney Docket No.: 08061.0057-00304 with functional magnetic resonance imaging (fMRI).
- the whole brain or at least one area of the brain for example, cortical gray matter (i.e., cortex), lateral ventricles, frontal lobe, parietal lobe, temporal lobe, occipital lobe, cingulate cortex, amygdala, piriform cortex, entorhinal cortex, hippocampus, hippocampal CA3 (pyramidal neurons), and/or hippocampal dentate gyrus (granule cell neurons) can be analyzed by MRI.
- neurodegeneration is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline.
- the amount of p-tau is reduced within 24 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, neurodegeneration is reduced within 12 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. In some embodiments, neurodegeneration is reduced within 6 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline. [0218] In some embodiments, neurodegeneration is reduced or maintained for at least 30 days after administration of the first dose of lemborexant. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained for at least 30 days after administration of the first dose of lemborexant.
- neurodegeneration is reduced within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or 55 days of administration of the first dose of lemborexant.
- the amount of p-tau in a subject’s CSF is reduced for at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or 55 days after administration of the first dose of lemborexant.
- neurodegeneration is reduced or maintained within 30 days of administration of the first dose of lemborexant.
- the amount of p-tau in a subject’s CSF is reduced or maintained for at least 30 days after administration of the first dose of lemborexant. In some embodiments, neurodegeneration is reduced or maintained within 60 days of administration of the first dose of lemborexant. [0221] In some embodiments, neurodegeneration is reduced or maintained within 45 days of administration of the first dose of lemborexant. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained for at least 45 days after administration of the first dose of lemborexant.
- neurodegeneration is reduced or maintained within 60 days of administration of the first dose of lemborexant. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained for at least 60 days after administration of the first dose of lemborexant. [0223] In some embodiments, neurodegeneration is reduced or maintained for at least 120 days after administration of the first dose of lemborexant. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained for at least 120 days after administration of the first dose of lemborexant. [0224] In some embodiments, neurodegeneration is reduced or maintained for at least 180 days after administration of the first dose of lemborexant.
- the amount of p-tau in a subject’s CSF is reduced or maintained for at least 180 days after administration of the first dose of lemborexant.
- the effects of lemborexant on neurodegeneration or p-tau are apparent after a period of time, for example, after 3 months, after 6 months, or after 9 months of treatment.
- neurodegeneration begins to reduce after at least 6 months of treatment.
- neurodegeneration is reduced after at least 3 months of treatment, for Attorney Docket No.: 08061.0057-00304 example, after 6 months or after 9 months of treatment.
- the amount of p-tau in a subject’s CSF is reduced or maintained after a period of at least 3 months. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained after a period of at least 6 months after treatment or a period of at least 9 months of treatment. In some embodiments, neurodegeneration is reduced or maintained for at least 6 months after administration of the first dose of lemborexant. In some embodiments, the amount of p-tau in a subject’s CSF is reduced or maintained for at least 6 months after administration of the first dose of lemborexant. [0226] In some embodiments, neurodegeneration is reduced for at least 1 year after administration of the first dose of lemborexant.
- the amount of p-tau in a subject’s CSF is reduced for at least 1 year after administration of the first dose of lemborexant. In some embodiments, the amount of t-tau in a subject’s CSF is reduced for at least 1 year after administration of the first dose of lemborexant. In some embodiments, the amount of A ⁇ in a subject’s CSF is reduced for at least 1 year after administration of the first dose of lemborexant. In some embodiments, the amount of fibrillar plaques is reduced for at least 1 year after administration of the first dose of lemborexant. In some embodiments, the amount of activated microglia cells is increased for at least 1 year after administration of the first dose of lemborexant. A.
- altering neurodegeneration e.g., reducing or delaying neurodegeneration, or slowing a rate of growth thereof
- a method of altering neurodegeneration comprising administering to the subject a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, Attorney Docket No.: 08061.0057-00304 wherein the therapeutically effective amount is sufficient to alter neurodegeneration in the subject.
- altering neurodegeneration comprises slowing progression of, slowing a rate of progression of, delaying progression of, arresting development of, and reversing progression of neurodegeneration.
- altering neurodegeneration comprises preventing the onset of or preventing development of pathology resulting from or coinciding with neurodegeneration). Altering neurodegeneration may reduce, delay, or slow a rate of onset and/or progression of symptoms of this pathology. In some embodiments, altering neurodegeneration comprises relieving or ameliorating one or more symptoms of neurodegeneration and/or improving one or more clinical metrics of pathology resulting from or coinciding with neurodegeneration (e.g., cognitive function, brain amyloid or tau levels, and/or expression of a biomarker). In some embodiments, altering neurodegeneration comprises preventing occurrence or reoccurrence of one or more symptoms of neurodegeneration. [0229] In some embodiments, the subject is amyloid negative.
- the subject may have mild cognitive impairment or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment.
- the neurodegeneration is altered relative to a reference. Accordingly, altering neurodegeneration may comprise reducing and/or delaying neurodegeneration, and/or slowing the rate thereof, as compared to a reference.
- the reference is a baseline measurement from the subject prior to treatment. In some embodiments, the reference is a baseline measurement from a control subject. The reference may be a measurement obtained from more than one control subjects, which is used as a standard or Attorney Docket No.: 08061.0057-00304 threshold measurement. In some embodiments, the reference is a measurement from a control subject administered a placebo.
- neurodegeneration is characterized by at least one of a loss of cortical thickness or a reduction in hippocampal volume. In some embodiments, altering neurodegeneration comprises maintaining or slowing a reduction of cortical thickness in the subject. In some embodiments, neurodegeneration is characterized by at least one of loss of a pyramidal neurons in the cortex or a loss of pyramidal or granule neurons in the hippocampus. In some embodiments, altering neurodegeneration comprises maintaining or slowing a reduction of hippocampal volume in the subject. In some embodiments, altering neurodegeneration comprises maintaining or reducing loss of pyramidal neurons or granule neurons. In some embodiments, altering neurodegeneration comprises reducing a rate of neurodegeneration.
- altering neurodegeneration comprises altering a neurofilament light chain (NfL) level.
- NfL levels may be altered in the blood and/or CSF of the subject.
- a further aspect of the present disclosure relates to a method of selecting a subject having Alzheimer’s disease (AD) or at risk of developing AD for treatment with lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, comprising: (a) obtaining from the subject a measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglial response, and biomarker expression; (b) comparing the measurement from the subject to a measurement from a reference; and (c) selecting the subject for treatment with lemborexant if the measurement from the subject differs from the measurement from the reference.
- AD Alzheimer’s disease
- a pharmaceutically acceptable salt thereof or a solvate thereof
- the subject has mild cognitive impairment or mild dementia. In some embodiments, the subject does not show signs of dementia and/or cognitive impairment. [0234] In some embodiments, the subject is at risk for further A ⁇ accumulation. The subject may be at risk for spread of tau pathology. The subject may be at risk for cognitive decline. In some embodiments, the subject may have intermediate levels of amyloid PET (e.g., approximately 20-40 centiloids). In some embodiments, the subject may have elevated levels of amyloid PET (e.g., > 40 centiloids). In some embodiments, the subject may be an ApoE4 carrier.
- the subject may have one or more risk factors for developing AD, such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease.
- the subject has early-stage AD.
- the subject has pre-AD.
- the subject has been diagnosed with AD, based on brain imaging, cognitive function, and/or biomarker criteria.
- obtaining at least one measurement comprises obtaining data from a brain scan of the subject and/or obtaining data from a biological sample from the subject.
- the data from the brain scan may indicate a level of tau phosphorylation, tau aggregation, A ⁇ plaque burden, and/or microglial response.
- the biological sample is a body fluid.
- the body fluid is cerebrospinal fluid (CSF), blood, or saliva.
- CSF cerebrospinal fluid
- the reference is a control.
- the reference is a baseline measurement from a control subject.
- the reference may be a measurement obtained from more than one control subjects, which is used as a standard or threshold measurement.
- the reference is a measurement from a control subject administered a placebo.
- the control does not have AD.
- the measurement from the subject may be higher than the measurement from the control who does not have AD.
- the measurement from the subject may be lower than the measurement from the control who does not have AD.
- the control has AD.
- the measurement from the subject is comparable to or higher than the measurement from the control who has AD.
- the measurement from the subject may be comparable to or lower than the measurement from the control who has AD.
- the measurement of tau phosphorylation comprises a measurement of phosphorylation on one more of T181, T217, S202, or S205.
- the measurement of tau aggregation comprises a measurement of insoluble tau aggregates (e.g., neurofibrillary tangles (NFTs)).
- the measurement of neurodegeneration comprises a measurement of cortical thickness and/or hippocampal volume or a measurement of loss of pyramidal neurons or granule neurons.
- the measurement of A ⁇ plaque burden comprises a measurement of A ⁇ plaque volume and/or growth of A ⁇ plaque volume.
- the measurement of A ⁇ plaque burden comprises a measurement of amyloid PET signal in a brain region of the subject or a measurement of A ⁇ in the CSF of the subject.
- the measurement of microglial response is a change in the expression of at least one microglial marker.
- the microglial marker may be Iba1, Clec7a, CD68, TMEM119, or P2RY12.
- the measurement of microglial response is a measurement of phagocytosis by microglia. IX.
- One aspect of the present disclosure relates to a method of monitoring treatment efficacy in a subject having Alzheimer’s disease (AD) or at risk for developing AD, comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (b) administering to the subject a dose of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; and (d) comparing the second measurement from the subject to the first measurement from the subject, wherein
- Another aspect of the present disclosure relates to a method of treating a subject having Alzheimer’s disease (AD) or at risk for developing AD, Attorney Docket No.: 08061.0057-00304 comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglial response, and biomarker expression; (b) administering to the subject a first dose of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (d) comparing the second measurement from the subject to the first measurement, and (d) administering a second dose of lemborexant if the first measurement differs from the second measurement in the comparison measurement.
- the first and second measurements may differ in that the second measurement is higher than the first.
- the second measurement may be lower than the first.
- the first measurement of microglial response may be higher than the second measurement of microglial response.
- Still a further aspect of the present disclosure relates to a method of monitoring treatment efficacy in a subject having Alzheimer’s disease (AD) or at risk for developing AD, comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (b) administering to the subject a dose of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; and (d) comparing the second measurement from the subject to the first measurement from the subject to obtain a comparison measurement, wherein a difference between the first measurement and Attorney Docket No.: 08061.0057-00304 the second measurement in the comparison measurement or a difference between the comparison measurement and a reference measurement indicates effective treatment with le
- Another aspect of the present disclosure relates to a method of treating a subject having Alzheimer’s disease (AD) or at risk for developing AD, comprising: (a) obtaining from the subject a first measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglial response, and biomarker expression; (b) administering to the subject a first dose of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof; (c) obtaining from the subject a second measurement of at least one of tau phosphorylation, tau aggregation, neurodegeneration, A ⁇ plaque burden, microglia function, and biomarker expression; (d) comparing the second measurement from the subject to the first measurement from the subject to obtain a comparison measurement, and (e) administering a second dose of lemborexant if the first measurement differs from the second measurement in the comparison measurement, or if the comparison measurement differs from a reference measurement.
- AD Alzheimer’s disease
- obtaining at least one measurement comprises obtaining data from a brain scan of the subject and/or obtaining data from a biological sample from the subject.
- the data from the brain scan indicates a level of tau phosphorylation, tau aggregation, A ⁇ plaque burden, and/or microglial response.
- the biological sample is a body fluid.
- the body fluid is cerebrospinal fluid (CSF), blood, or saliva.
- CSF cerebrospinal fluid
- the first measurement from the subject is higher than the second measurement from the subject. In some embodiments, the first measurement from the subject is lower than the second measurement from the subject.
- the reference measurement is obtained from at least one control.
- the reference measurement is a comparison of a first measurement from a control and a second measurement from a control.
- the comparison measurement is higher than the reference measurement.
- the comparison measurement is lower than the reference measurement.
- the comparison measurement which compares a first measurement from the subject and a second measurement from the subject, may indicate that no change has occurred.
- the reference measurement meanwhile, may indicate that a change occurred in a control.
- the difference between the comparison measurement and the reference measurement may indicate whether a treatment has been efficacious and/or whether a second dose of lemborexant should be administered.
- the reference measurement is a measurement from a control.
- the reference measurement may be obtained from more than one control subjects, which is used as a standard or threshold measurement.
- the reference measurement is a measurement from a control subject administered a placebo.
- the control does not have AD.
- the comparison measurement is higher than the reference Attorney Docket No.: 08061.0057-00304 measurement.
- the comparison measurement is lower than the reference measurement.
- the control has AD, for example, untreated AD.
- the comparison measurement is higher than the reference measurement. In some embodiments, the comparison measurement is lower than the reference measurement.
- the measurement of tau phosphorylation comprises a measurement of phosphorylation of one or more of T181, T217, S202, S205, or T231.
- the measurement of tau aggregation comprises a measurement of insoluble tau aggregates (e.g., neurofibrillary tangles (NFTs)).
- the measurement of neurodegeneration comprises a measurement of cortical thickness and/or hippocampal volume or a measurement of loss of pyramidal neurons or granule neurons.
- the measurement of A ⁇ plaque burden comprises a measurement of A ⁇ plaque volume and/or growth of A ⁇ plaque volume.
- the measurement of A ⁇ plaque burden comprises a measurement of amyloid PET signal in a brain region of the subject or a measurement of A ⁇ in the CSF of the subject.
- the measurement of microglial response is a measure of expression of at least one microglial marker.
- the microglial marker may be Iba1, Clec71, P2RY12 or TMEM 119.
- the measurement of microglial response is a measurement of phagocytosis by microglia.
- the measurement of a biomarker expression is a measurement of Ifnb1, MMP2, and/or Bace1 expression.
- the subject is amyloid-negative.
- the subject has A ⁇ plaques.
- the subject has mild cognitive impairment or mild dementia.
- the subject does not show signs of dementia and/or cognitive impairment.
- the subject is at risk for further A ⁇ accumulation.
- the subject may be at risk for spread of tau pathology.
- the subject may be at risk for cognitive decline.
- the subject may have intermediate levels of amyloid PET (e.g., approximately 20-40 centiloids). In some embodiments, the subject may have elevated levels of amyloid PET (e.g., > 40 centiloids). In some embodiments, the subject may be an ApoE4 carrier. In some embodiments, the subject may have one or more risk factors for developing AD, such as having a family history with a first degree relative having AD or dementia, having an age of 65 years or older, being female, having or recovering from traumatic brain injury, and suffering from other conditions such as obesity, diabetes, heart disease, and/or blood vessel disease. [0269] In some embodiments, wherein the subject has early-stage AD. In some embodiments, the subject has pre-AD.
- a dose of lemborexant may refer to a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt Attorney Docket No.: 08061.0057-00304 thereof, or a solvate thereof.
- the methods disclosed herein comprise administering orally 5 mg to 20 mg of lemborexant to the subject once per day. In some embodiments, one dose of 20 mg of lemborexant is administered to the subject once per day. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day.
- one dose of 25 mg of lemborexant is administered to the subject once per day for at least 2 days. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day for at least 5 days. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day for at least one week. In some embodiments, one dose of 25 mg of lemborexant is administered to the subject once per day.
- one dose of 25 mg of lemborexant is administered to the subject once per day for at least one month.
- the methods disclosed herein comprise administering orally a dosage form comprising lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 10 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 15 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 20 mg of lemborexant to the subject.
- the methods disclosed herein comprise administering orally a dosage form comprising 25 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise Attorney Docket No.: 08061.0057-00304 administering orally a dosage form comprising 30 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 35 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 40 mg of lemborexant to the subject. In some embodiments, the methods disclosed herein comprise administering orally a dosage form comprising 45 mg of lemborexant to the subject.
- the methods disclosed herein comprise administering orally a dosage form comprising 50 mg of lemborexant to the subject.
- Dosage forms of the present disclosure comprise lemborexant in a therapeutically effective amount for treatment of when administered in accordance with the teachings of the present disclosure.
- the unit dose of the effective amount in a dosage form is from 0.5 mg to 100 mg, from 2 mg to 75 mg, from 2 mg to 70 mg, from 2 mg to 65 mg, from 2 mg to 60 mg, from 2 mg to 55 mg, from 2 mg to 50 mg, from 2 mg to 45 mg, from 2 mg to 40 mg, from 2 mg to 35 mg, from 2 mg to 30 mg, from 2 mg to 25 mg, from 2 mg to 20 mg, from 2 mg to 15 mg, from 2 mg to 15 mg, 2 mg, 2.5 mg, 4 mg, 5 mg, 8 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, or 50 mg.
- the unit dose is not limited by the type of the dosage form or the number of dosage forms for single dose.
- the unit dose may be 2.5 mg. In some embodiments, the unit dose may be 5 mg. In some embodiments, the unit dose may be 10 mg. In some embodiments, the unit dose may be 7.5 mg. In some embodiments, the unit dose may be 12.5 mg. In some embodiments, the unit dose may be 15 mg. In some embodiments, the unit dose may be 18 mg. In some embodiments, the unit dose may be 20 mg. In some embodiments, the unit dose may be 22 mg. In some embodiments, the unit dose Attorney Docket No.: 08061.0057-00304 may be 25 mg. In some embodiments, the unit dose may be 30 mg. In some embodiments, the unit dose may be 32 mg. In some embodiments, the unit dose may be 35 mg.
- the unit dose may be 40 mg. In some embodiments, the unit dose may be 50 mg.
- a therapeutically effective amount of lemborexant, a pharmaceutically acceptable salt thereof, or a solvate thereof, as administered herein may comprise a dosage falling within a range, for example, a range of 5 mg to 50 mg per day.
- the therapeutically effective amount of lemborexant administered to the subject is in a range of 5 mg to 50 mg per day.
- the therapeutically effective amount of lemborexant administered to the subject may be in a range of 10 mg to 30 mg per day.
- the therapeutically effective amount of lemborexant administered to the subject is selected from 5 mg, 7.5 mg, 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg and 30 mg per day. [0275] In some embodiments, the therapeutically effective amount of lemborexant administered to the subject is 20-25 mg per day. [0276] In some embodiments, one dose of 20 mg of lemborexant is administered to the subject once per day. [0277] In some embodiments, lemborexant is administered at a first dose for a first period, a second dose for a second period, and optionally, at a third dose for a third period.
- Each of the first period, the second period, and the third period may be 1 week.
- the first dose is lower than the second dose, and optionally, the second dose is lower than the third dose.
- the first dose is 5 mg of lemborexant once per day
- the second dose is 10 mg of lemborexant once per day
- the third dose is 20-25 mg of lemborexant once per day.
- the first dose is 5 mg or 7.5 mg lemborexant once per day
- the second dose is 10 mg, 12.5 mg, 15 mg, or 17.5 mg of lemborexant once per day
- the third dose is 20 mg, 22.5 mg, 25 mg, 27.5 mg or 30 mg of lemborexant once per day.
- the first dose is higher than the second dose, and optionally, the second dose is higher than the third dose.
- the first dose is 20-25 mg of lemborexant once per day
- the second dose is 10 mg of lemborexant once per day
- the third dose is 5 mg of lemborexant once per day.
- the first dose is 20 mg, 22.5 mg, 25 mg, 27.5 mg or 30 mg of lemborexant once per day
- the second dose is 10 mg, 12.5 mg, 15 mg, or 17.5 mg of lemborexant once per day
- the third dose is 5 mg or 7.5 mg of lemborexant once per day.
- lemborexant may be administered to the subject over a period of time.
- the methods described herein comprise administering lemborexant to the subject for at least 6 months.
- the methods described herein comprise administering lemborexant to the subject for at least 9 months, at least 12 months, or at least 15 months.
- the methods described herein comprise administering lemborexant to the subject for at least 18 months. In some embodiments, the methods described herein comprise administering lemborexant to the subject for at least 24 months, 30 months, or 36 months. In some embodiments, lemborexant may be administered for the remainder of the subject’s life. Attorney Docket No.: 08061.0057-00304 XI.
- Pharmaceutical Compositions [0281] In some embodiments, a dosage form of the present disclosure may constitute one or more pharmaceutical compositions comprising lemborexant together with pharmaceutically acceptable excipients.
- composition used herein includes a product comprising a particular ingredient in a particular amount and any product directly or indirectly brought about by the combination of particular ingredients in particular amounts.
- a term related to the pharmaceutical composition is intended to include a product comprising an active ingredient and an inert ingredient constituting a carrier and include every product directly or indirectly brought about by the combination, complexation or aggregation of any two or more ingredients or the dissociation, other kinds of reactions or interaction of one or more ingredients.
- the pharmaceutical composition of the present disclosure includes every composition prepared by mixing the compound of the present disclosure with a pharmaceutically acceptable carrier.
- the term “pharmaceutically acceptable” means that a carrier, diluent, excipient, or vehicle is compatible with other components of a formulation and is nontoxic to a subject.
- Solid dosage forms of the present disclosure include capsules, granules, lozenges, pellets, pills, powders, suspensions, and tablets.
- the pharmaceutical compositions of the present disclosure may be prepared using standard techniques and manufacturing processes generally known in the art. See, e.g., the monograph of Japanese Pharmacopoeia, 16th Edition; and Pharmaceutical Dosage Forms of U.S. Pharmacopoeia-NF, Chapter 1151.
- compositions comprise lemborexant.
- pharmaceutical compositions further comprise at least one additional component chosen from pharmaceutically acceptable carriers, pharmaceutically acceptable vehicles, and pharmaceutically acceptable excipients.
- the at least one additional component in the pharmaceutical compositions is chosen depending upon the route of administration for which the pharmaceutical composition is intended.
- suitable routes of administration for which the pharmaceutical composition may be used include parenteral, oral, inhalation spray, topical, rectal, nasal, buccal, vaginal and implanted reservoir administration.
- parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intracisternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
- the mode of administration is chosen from intravenous, oral, subcutaneous, and intramuscular administration.
- Sterile injectable forms of the compositions of this disclosure may be, for example, aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents known in the art.
- the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
- a nontoxic parenterally acceptable diluent or solvent for example, as a solution in 1,3-butanediol.
- vehicles and solvents that may be employed include water, Ringer's solution, and isotonic sodium chloride solution.
- sterile, fixed oils may be employed as a solvent and/or suspending medium.
- any bland fixed oil may be employed including synthetic mono- or di-glycerides.
- Fatty acids such as oleic acid and its glyceride Attorney Docket No.: 08061.0057-00304 derivatives are useful in the preparation of injectables, as are natural pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
- oils such as olive oil or castor oil, especially in their polyoxyethylated versions.
- These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
- lemborexant may be provided in an acceptable oral dosage form, including, but not limited to, suspensions, capsules, tablets, oral disintegrating tablets, sprinkles, and other oral formulations that would be easy to swallow.
- lemborexant is provided in the form of tablet or capsules.
- lemborexant is provided in the form of crushable tablets.
- carriers commonly used include lactose and cornstarch.
- Lubricating agents such as magnesium stearate, may also be added.
- useful diluents include lactose and dried cornstarch.
- the active ingredient is combined with an emulsifying and/or suspending agent.
- certain sweetening, flavoring or coloring agents may also be added.
- a method of reducing or maintaining the amount of p-tau or t-tau in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- any one of embodiments 1-15 wherein the amount of p-tau is reduced within 48 hours of administration of the first dose of lemborexant to the subject relative to the subject’s baseline.
- a method of reducing neurodegeneration in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- Attorney Docket No.: 08061.0057-00304 18.
- the method of embodiment 17, wherein the reduction of neurodegeneration is observed by maintenance or slowing of reduction of the thickness of the cortex relative to a subject’s baseline or to a subject to whom placebo was administered. 19.
- a method of increasing the number of activated microglial cells in a subject comprising administering to a subject in need thereof a therapeutically effective amount of lemborexant.
- 34 The method of embodiment 33, wherein the number of activated microglial cells is increased relative to baseline.
- Attorney Docket No.: 08061.0057-00304 35 The method of any one of embodiments 33 or 34, wherein the activated microglial cells are phagocytic microglial cells.
- any one of embodiments 33-36 wherein the whole brain or at least one area of the brain is analyzed by PET. 38.
- EXAMPLES Example 1 Clinical Study Protocols a.
- Trial 1 Acute Effects of Lemborexant on CSF Amyloid-Beta and Tau
- Inclusion criteria will be: o Age 60-80 years o Any sex o Any race/ethnicity o Mini-Mental Status Examination score (MMSE) ⁇ 27 o Positive plasma A ⁇ test (i.e., amyloid-positive) o Pittsburgh Sleep Quality Index >5 Attorney Docket No.: 08061.0057-00304
- Exclusion Criteria will be: Cognitive impairment as determined by history of MMSE ⁇ 27 Inability to speak or understand English Any sleep disorders other than insomnia ⁇ No history of moderate-to-severe sleep-disordered breathing and STOP-Bang score > 5 ⁇ History or reported symptoms suggestive of restless legs syndrome, narcolepsy or other sleep disorders ⁇ No more than mild sleep apnea (AHI ⁇ 16) on P
- Procedure Randomized participants will be admitted in the early afternoon (Night 1). All participants will have their sleep monitored with unattended full-montage PSG (TrackIt TM ; Lifelines, Troy, IL) that will allow for sleep staging according to the gold standard American Academy of Sleep Medicine criteria and has already been used in similar studies to monitor sleep for 36-48 hours.
- a lumbar catheter and two IVs will be placed in each participant for collecting 6 ml of CSF every 2 hours for 48 hours.
- the lumbar catheter ports will be placed on the outside of the gown sleeve for easy access to minimize disturbance during fluid collection.
- Sampling start time will begin ⁇ 1 hour prior to the typical bedtime defined by sleep logs for each participant in order to allow Attorney Docket No.: 08061.0057-00304 for 13 C6-leucine infusion and frequent sampling of blood prior to bedtime.
- Six milliliters of blood will be collected at the following time points: 0 (baseline), 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 26 hours, 28 hours, 30 hours, 32 hours, 34 hours, 36 hours, 38 hours, 40 hours, 42 hours, 44 hours, 46 hours, and 48 hours (Table 1). Table 1.
- CSF tau, phosphorylated tau, and A ⁇ kinetics will be quantified by mass spectrometry.
- Phosphorylated tau ratios (pT181/T181, pS202/S202, pT217/T217, p- tau/t-tau) will be normalized to the first time point (hour 0).
- Unstructured covariance matrix will be used and if there is convergence issue, various other covariance matrix structure (e.g., compound symmetry, First ⁇ order autoregressive) will be compared and the best fit structure will be selected for final analysis based on Akaike information criterion (AIC).
- AIC Akaike information criterion
- Trial 2 Treatment of Subjects Having Preclinical or Early Alzheimer’s Disease [0302] This trial will evaluate the efficacy of lemborexant in the prevention or delay of A ⁇ accumulation, spread of downstream tau pathology, and cognitive decline across the continuum of preclinical and early AD.
- Amyloid-ß (Aß) accumulation often begins more than a decade prior to the clinical stages of Alzheimer’s disease (AD) and is thought to play a critical role in accelerating the spread of tauopathy and neurodegeneration during the preclinical stages of the disease. Multiple neuroimaging and biomarker observational studies demonstrate that A ⁇ accumulation is associated with increased risk of cognitive decline among clinically normal older individuals.
- the trial will utilize NAV4694 (flutafuranol) amyloid PET imaging to assess fibrillar amyloid pathology for eligibility and longitudinal outcomes, and the MK6240 tau PET tracer to assess spread of neurofibrillary tangle and tau neurite pathology longitudinally.
- Clinical outcomes include the Preclinical Alzheimer Cognitive Composite-5 (PACC-5) composed of the Free and Cued Selective Reminding Test, Paragraph Recall IIa, Digit-Symbol, MMSE, and Semantic Category Fluency, as well as the Cognitive Function Index (CFI), a participant- and study- partner report of cognitive function.
- PACC-5 Preclinical Alzheimer Cognitive Composite-5
- Paragraph Recall IIa Digit-Symbol
- MMSE Digit-Symbol
- CFI Cognitive Function Index
- scans of the patient’s cortex and hippocampus will be performed to assess extent of neurodegeneration.
- CSF and blood will be collected to measure AD biomarkers such as A ⁇ , tau, p-tau, and NfL. i.
- Group 1 Patients with Preclinical Alzheimer’s Disease – intermediate levels of amyloid
- the goal of this trial is to determine if lemborexant treatment leads to primary prevention or delay of AD, through preventing or slowing early A ⁇ build-up in Attorney Docket No.: 08061.0057-00304 the brain.
- the trial will enroll cognitively-normal individuals with intermediate levels of amyloid on screening PET imaging (approximately 20-40 centiloids), thought to be in the earliest preclinical stages of AD who are at risk for further A ⁇ accumulation and early spread of tau pathology over four years.
- Patient Inclusion Criteria are provided.
- MMSE Mini-Mental State Examination
- CDR clinical dementia rating
- Patients will have intermediate amyloid levels on PET imaging (20-40 centiloids).
- Lemborexant administration Lemborexant or matched placebo will be administered to participants every month during in-person study visits where treatment adherence and adverse events will be assessed.
- Lemborexant is approved by the FDA for the treatment of insomnia in doses from 5-10 mg. After an interim analysis for safety and efficacy, the lemborexant dose may be increased to 20 mg.
- Primary outcomes the primary outcome measure of the trial is amyloid PET SUVr at 6 months, measured and compared to placebo.
- Biomarker outcomes will include tau PET, measurements of A ⁇ , phospho-tau, and protofibrils in CSF and plasma.
- Secondary outcomes tau PET will be measured.
- Exploratory outcomes: in CSF, the biomarkers A ⁇ , tau, phospho-tau, neurogranin (NG), neurofilament light chain (NfL) will be measured. In plasma, NfL, phospho-tau 181, and phospho-tau 217 will be measured.
- Clinical outcomes to measure cognition will include tests to obtain a Preclinical Alzheimer’s Disease Cognitive Composite 5 (PACC5) scale for cognition and a Cognitive Functional Index (CFI).
- PACC5 Preclinical Alzheimer’s Disease Cognitive Composite 5
- CFI Cognitive Functional Index
- ii. Preclinical Alzheimer’s Disease – elevated levels of amyloid The goal of this trial is to determine if lemborexant treatment leads to primary prevention or delay of AD, through preventing early A ⁇ build-up in the brain.
- Attorney Docket No.: 08061.0057-00304 The trial will enroll cognitively-normal individuals with elevated levels of amyloid on screening PET imaging (approximately >40 centiloids), who are at high risk for cognitive decline over four years.
- Patient Inclusion Criteria Patient Inclusion Criteria.
- MMSE Mini-Mental State Examination
- CDR global clinical dementia rating
- Patients will have elevated amyloid PET levels >40.
- Lemborexant administration Lemborexant or matched placebo will be administered to participants every month during in-person study visits where treatment adherence and adverse events will be assessed.
- Lemborexant is approved by the FDA for the treatment of insomnia in doses from 5-10 mg. After an interim analysis for safety and efficacy, the lemborexant dose may be increased to 20 mg.
- Primary outcomes To test the effects of lemborexant on cognitive decline in patients with elevated amyloid, the primary outcome measure of the trial is the Preclinical AD Cognitive Composite 5 (PACC5) at 6 months. Biomarker outcomes will include tau PET, measurements of A ⁇ , phospho-tau, and protofibrils in CSF and plasma. [0322] Second outcomes. The Cognitive Function Index (CFI) will be measured. Amyloid PET and tau PET will be measured. [0323] Exploratory outcomes. Clinical measurements will be ADCS ADL- prevention, Computerized Cognitive Composite, ISLT, Trails, CDR-SB, and time to CDR 0.5. vMRI and rs-MRI will be determined.
- PACC5 Preclinical AD Cognitive Composite 5
- CFI Cognitive Function Index
- Example 2 Mouse Study of Tau-Mediated Neurodegeneration [0325] P301S/E4 and E4 knock-in non-tau depositing mice were orally gavaged with 30mg/kg lemborexant or vehicle daily from 7.5 months of age, when tau-mediated neuroinflammation is observed without overt neuronal loss, until 9.5 months of age (Fig.2a). A.
- Atrophy of the hippocampus and piriform/entorhinal cortex was significantly ameliorated by approximately 50% concomitant with less enlargements of the lateral ventricles.
- the granule cell and pyramidal cell neuronal layers were visibly thicker in lemborexant-treated P301S/E4 mice (FIG.7G – FIG. 7K), which was validated by reduced plasma neurofilament light chain levels (FIG. 3J), evidencing a robust improvement in neuronal damage and degeneration.
- Microglial reactivity [0331] To investigate whether the marked reduction in tau-mediated neurodegeneration in lemborexant-treated mice was linked with reduced microglial reactivity, different markers of reactive microglia across the spectrum of disease- associated or homeostatic populations in both hippocampus and piriform cortex were quantified. Substantial changes, predominantly in the hippocampus and most notably in the CA3 regions (FIG.4), were observed (FIG.4).
- RNA sequencing in bulk hippocampal tissue was performed.
- genes such as Adra2b, Trh, Trhr2, Mpzl2, Slc22a6, Pla2g2f, Ptgdr, Foxp2 are genes that modulate sleep. More specifically, thyrotropin-releasing hormone (Trh) and its receptor (Trhr2) modulate behavioral arousal, in part, through orexin.
- TRH application transforms GABAergic neurons from the burst-firing mode typically associated with the synchronized cortical activity that occurs during NREM sleep to the tonic, single-spike mode of action potential generation associated with desynchronized cortical activity that occurs during wakefulness and REM sleep.
- Trh and Trhr2 suggest that DORA-induced NREM sleep further interacts with humoral regulation of sleep-wake behavior to promote sleep, particularly in the presence of tau, as these effects were absent in non-tau E4 mice.
- Slc22a6 Pla2g2f, Ptgdr expression that modulate sleep-wake through potent endogenous somnogens such as prostaglandins.
- phospholipase A2 play a major role in cell growth differentiation and inflammation, and are associated with metabolic changes in patients with obstructive sleep apnea.
- PS19 tau transgenic mice harboring 1N4R tau and overexpressing human P301S tau mutation were used24. These mice have been backcrossed to C57BL/6 for more than ten generations. Human apoE4 knock-in mice were generated as described in Mol. Neurodegener.14, (2019) and crossed to P301S mice for several generations to produce experimental P301S/E4 mice. Littermates of same sex were randomly assigned to experimental groups. Only male animals were used and sacrificed at 9.5 months of age. All mice were housed in specific pathogen-free conditions and under Attorney Docket No.: 08061.0057-00304 the same 12h light/dark cycle, ambient room temperature as well as with food and water available ad libitum.
- mice were gavaged daily with a single 30mg/kg dose of lemborexant or 0.5% methylcellulose vehicle at ZT13, one hour post dark onset, starting at 7.5M of age until euthanizing at 9.5M of age.
- Tissue collection All mice were perfused between ZT3 and ZT7, at a time window when mice were sleep deprived, to avoid circadian influence on transcriptional fluctuation of microglial gene expression. Prior to transcardiac perfusion, mice were anesthetized with pentobarbital (50 mg/kg, intraperitoneal). Blood was collected from the heart before transcardiac perfusion, which was centrifuged at 5000xg for 5mins at 4°C to obtain plasma.
- mice were transcardially perfused with ice-cold phosphate-buffered saline containing 0.3% heparin.
- One hemibrain was dissected, snap-frozen and stored at ⁇ 80°C for biochemical analyses.
- the other hemibrain was immerse-fixed for 24h in 4% paraformaldehyde following by cryoprotection in 30% sucrose for 48h and frozen at ⁇ 80°C until tissue samples were sectioned for immunohistochemical analyses.
- Measurement and analysis of sleep-wake states Sleep-wake behavior in mice were monitored using electroencephalography (EEG) and independently using PiezoSleep mouse behavioral tracking system (SignalSolutions).
- mice were anesthetized with isofluorane (0.5-3%). Any signs of pain were assessed by toe pinching before an incision was made. Mice were then surgically implanted with screw electrodes in the skull for EEG and stainless wire electrodes in the nuchal muscle for electromyography (EMG). After a midline vertical incision to expose the skull, forceps and 3% hydrogen Attorney Docket No.: 08061.0057-00304 peroxide were used to remove any connective tissue and dry the skull for electrode placement.
- Burr holes for the frontal reference electrodes were made (anterior +0.5 ⁇ mm, lateral ⁇ 0.5 ⁇ mm; bregma) using a micro drill with a 0.9 ⁇ mm tip and screws were secured in the skull.
- Two bilateral active recording electrodes were placed over the parietal cortex (posterior ⁇ 2.5 ⁇ mm, lateral ⁇ 1.5; bregma) and a ground screw secured over the cerebellum (posterior ⁇ 6.2 ⁇ mm, lateral ⁇ 0.5; bregma), using the same technique as the reference electrode.
- the exposed skull, screws, and all wires were covered in a layer of dental cement (SNAP, Parkell) with the pin header secured to the head for subsequent recording.
- mice were placed in a warmed chamber to fully recover from anesthesia and individually housed in monitoring cages with fresh bedding, water, food and Carprofen (orally; 1 ⁇ 4 tab of 5g tab; ad libitum) supplement. After recovery from surgery over three days, mice were habituated in the recording cage for two weeks, followed by undisturbed EEG/EMG recording performed for two consecutive days in freely moving mice. Bilateral cortical EEG signals were acquired by a P511K A.C.
- Preamplifier (Grass ⁇ Telefactor Instruments, Warwick, RI USA), digitized with a BIOPAC MP150, recorded digitally using the BIOPAC's AcqKnowlege software with a sampling rate of 250Hz, and converted into (.edf) format for analysis.
- EEG was processed in MATLAB (MathWorks) through a band-pass filter of 1-30 Hz to remove DC offset and high- frequency noise.
- EEG/EMG recordings were manually scored in 10-second epochs for wake, NREM and REM sleep to create a calibration file containing mixture-z scoring variables specific for the recording subject.
- the calibration file was imported Attorney Docket No.: 08061.0057-00304 into AccuSleep, a machine learning-based, automated sleep scoring program in MATLAB, to complete the remainder of the scoring.
- PiezoSleep mouse behavioral tracking system (Signal Solutions, LLC, Lexington, KY, USA) was used.
- the non-invasive method includes a thin dielectric piezo sensor pad that generates a voltage signal in response to changes in real-time fluctuations in pressure on its surface. Mice were individually housed with the piezo pad underneath fresh bedding, with fresh water and food available ad libitum and recorded without disturbance over a period of six days. Data was acquired using SleepStats software (Signal Solutions, LLC, Lexington, KY, USA).
- volumetric analysis Volumetric analysis of the hippocampus, entorhinal/piriform cortex, and ventricle was performed via stereological methods by assessing sections spaced by 180 ⁇ m starting from bregma ⁇ 1.3 mm to bregma ⁇ 3.1 mm (16 – 18 sections per mouse depending on the severity of brain atrophy).30 ⁇ m microtome-cut sections mounted on slides were briefly immersed in distilled water before incubating in pre-warmed 0.1% cresyl violet at 37°C for six minutes. Following this, tissues were rinsed in distilled water and transferred to 70%, 95% and 100% ethanol sequentially, for two minutes each.
- Primary antibodies were diluted in blocking buffer and incubated at 4°C overnight with slow agitation unless stated otherwise. Primary antibodies were used as follows: IBA1 (1:500; 019-19741, Fujifilm or NB100-1028, Novus Biologicals), CD68 (1:00; FA-11, BioRad), P2RY12 (1:100 at room temperature, HPA013796, Sigma-Aldrich), TMEM119 (1:500; E3E1O, Cell Signaling Technology), Clec7a (1:50 at room temperature; mabg-mdect, InvivoGen), GFAP (1:2000; 2E1.E9 Alexa Flour 488- conjugated, BioLegend), APOE (1:300; D7I9N, Cell Signalling), PSD-95 (1:200, 51- 6900, Thermo Fisher Scientific), VGLUT1 (1:200, AB5905, Merck Millipore).
- Frozen mouse hippocampal tissue was weighed and homogenized in a bullet blender homogenizer (Next Advance) using beaded tubes with 200 ⁇ l RAB buffer pH 7.0 (100mM MES, 1mM EGTA, 0.5mM MgSO4, 750mM NaCl, 20mM NaF, 1mM Na3VO4) supplemented with 1x protease inhibitor (cOmpleteTM, Roche) and 1x phosphatase inhibitor (PhosSTOP, Roche).
- RAB buffer pH 7.0 100mM MES, 1mM EGTA, 0.5mM MgSO4, 750mM NaCl, 20mM NaF, 1mM Na3VO4
- proteins were extracted with RIPA buffer pH 8.0 (150mM NaCl, 50mM TRIS, 0.5% deoxycholic acid, 1% Triton-X 100, 0.1% sodium deoxycholate, 5mM EDTA, 20mM NaF, 1mM Na3VO4) supplemented with protease and phosphatase inhibitors. After a five min clearing for RIPA insoluble material at 5000 ⁇ g at 4°C, the supernatant was again ultracentrifuged for 30 minutes at 50’000 ⁇ g to obtain the RIPA soluble protein fraction.
- RIPA buffer pH 8.0 150mM NaCl, 50mM TRIS, 0.5% deoxycholic acid, 1% Triton-X 100, 0.1% sodium deoxycholate, 5mM EDTA, 20mM NaF, 1mM Na3VO4
- the RIPA insoluble pellet was dissolved with ice-cold 70% formic acid (FA) and sonicated for one minute at 30% amplitude in short pulses at room temperature using sonicator (Model FB120, Fisher Scientific), followed by a final ultracentrifugation for 20 minutes at 50’000 ⁇ g at 4°C. Protein concentrations were measured for RIPA fractions using a BCA assay (Pierce). All samples were aliquoted and frozen at ⁇ 80°C until use. [0350] Tau ELISA: Human tau and pTau were measured in in RAB, RIPA and 70% FA fractions using a sandwich ELISA and normalized to tissue weight as described28.
- the coating antibodies for total human tau, and pTau were TAU-5 (mouse monoclonal, 20 ⁇ g/ml), and HJ14.5 (mouse monoclonal, 20 ⁇ g/ml), respectively.
- the capture antibodies for total human tau and pTau were HT7- biotinlyated (MN1000B, ThermoFisher Scientific) and AT8-biotinlyated (MN1020B, thermos Fisher Scientific), respectively.
- NFL concentration Plasma NFL concentration was measured with NF-Light Simoa Assay Advantage kit using Quanterix. The measurement was performed following the manufacturer’s instructions.
- RNA extraction Frozen hippocampal tissue was weighed and homogenized in RNAase-free beaded tubes (REDE, Next Advance) in chloroform with TRIzolTM. Samples were centrifuged for 15 minutes at 12’000xg at 4°C and the aqueous upper supernatant transferred for RNA isolation with the RNeasy Mini Kit Attorney Docket No.: 08061.0057-00304 (Qiagen) following manufacturer’s instructions. RNA quality was controlled using Bioanalyzer prior to Next Generation Sequencing by Clontech SMARTer. [0353] RNA sequencing and analyses: Samples were prepared according to library kit manufacturer’s protocol, indexed, pooled, and sequenced on an Illumina NovaSeq 6000.
- RNA-seq reads were then aligned to the Ensembl release 76 primary assembly with STAR version 2.7.9a (Doblin et al). Gene counts were derived from the number of uniquely aligned unambiguous reads by Subread:featureCount version 2.0.3. Isoform expression of known Ensembl transcripts were quantified with Salmon version 1.5.2. Sequencing performance was assessed for the total number of aligned reads, total number of uniquely aligned reads, and features detected.
- the ribosomal fraction, known junction saturation, and read distribution over known gene models were quantified with RSeQC version 4.0. All gene counts were then imported into the R/Bioconductor package EdgeR and TMM normalization size factors were calculated to adjust for samples for differences in library size. Ribosomal genes and genes not expressed in the smallest group size minus one sample greater than one count-per-million were excluded from further analysis. The TMM size factors and the matrix of counts were then imported into the R/Bioconductor package Limma.
- Weighted likelihoods based on the observed mean- variance relationship of every gene and sample were then calculated for all samples with the voomWithQualityWeights function and were fitted using a Limma generalized linear model with additional unknown latent effects as determined by surrogate variable analysis (SVA).
- SVA surrogate variable analysis
- the performance of all genes was assessed with plots of the residual standard deviation of every gene to their average log-count with Attorney Docket No.: 08061.0057-00304 a robustly fitted trend line of the residuals.
- Differential expression analysis was then performed to analyze for differences between conditions and the results were filtered for only those genes with Benjamini-Hochberg false-discovery rate adjusted p-values less than or equal to 0.05.
- Perturbed KEGG pathways where the observed log2 fold-changes of genes within the term were significantly perturbed in a single-direction versus background or in any direction compared to other genes within a given term with p-values less than or equal to 0.05 were rendered as annotated KEGG graphs with the R/Bioconductor package Pathview.
- the Limma voomWithQualityWeights transformed log2 counts-per-million expression data was then analyzed via weighted gene correlation network analysis with the R/Bioconductor package WGCNA. Briefly, all genes were correlated across each other by Pearson correlations and clustered by expression similarity into unsigned modules using a power threshold empirically determined from the data.
- Example 3 Effects of Lemborexant and Doxepin in a Model of A ⁇ Burden A. Sleep-Wake Behavior [0357] Changes in sleep were assessed in APPswe/PS1deltaE9 (also called “PSAPP”) mice after administration of either doxepin or lemborexant.
- FIG.9A shows a schematic of the experimental design and graphs of the effects of doxepin and lemborexant on total sleep (FIG.9B), light phase sleep (FIG.9C), and dark phase sleep (FIG.9D).
- FIG.9E shows the sleep percentage across time- of-day timepoints after drug injection.
- P values are from 2-way repeated measures ANOVA.
- the data indicate that lemborexant and doxepin each increase total sleep time in PSAPP mice, and the effects are comparable after treatment with each drug. Notably, DOX-induced sleep is spread throughout the day, whereas LEM induced sleep is limited to light phase (natural rest phase for mice).
- B. Amyloid Plaque Deposition [0360] Either lemborexant or doxepin were chronically administered to PSAPP mice in order to determine the long-term effects of each drug effects on amyloid plaque deposition.
- FIG.10A shows a schematic for the timing of treatment of PSAPP mice. Females develop plaques faster/younger, so males and females were staggered to allow combination of data.
- FIG.10B shows representative images of brain sections stained with X34, which labels fibrillar amyloid plaques.
- FIG.10C shows the quantification of plaque burden (% area X34 staining) in different brain regions (hippocampus, somatomotor cortex, somatosensory cortex, and piriform Attorney Docket No.: 08061.0057-00304 cortex). Error bars indicate mean ⁇ SEM, each dot is a mouse.
- FIG.11 shows that chronic lemborexant reduces total amyloid plaque burden in PSAPP mice more effectively than doxepin.
- FIG.11A shows brain sections stained for total amyloid plaques burden using the anti-A ⁇ antibody HJ3.4.
- FIG.11B shows the quantification of plaque burden (measured as the % area showing HJ3.4 staining) in different brain regions (hippocampus, somatomotor cortex, somatosensory cortex, and piriform cortex).
- FIG.12A shows representative Western blots, with beta-tubulin as Attorney Docket No.: 08061.0057-00304 a loading control.
- FIG.12B shows the quantification of band intensity. Error bars indicate mean ⁇ SEM, each dot is a mouse. P values from 1-way ANOVA are shown. [0366] The results show that neither lemborexant nor doxepin alter APP processing/cleavage in PSAPP mice. D.
- FIG.13A shows representative images from brain section obtained from PSAPP mice after administration of either lemborexant or doxepin.
- FIG.13B shows the quantification of plaque volume, indicating that similar sized plaques were quantified across conditions, and peri- plaque Iba1 volume.
- FIG.14A shows representative images of brain sections.
- FIG. Attorney Docket No.: 08061.0057-00304 14B shows the quantification of Iba1-colocalized CD68 around each plaque, obtained using Imaris software. Error bars indicate mean ⁇ SEM, each dot is the average of 8-10 plaques from a single mouse. P values from 1-way ANOVA are shown. [0370] The results indicate that the administration of lemborexant, but not doxepin, increases phagocytic subtypes of microglia surrounding fibrillar amyloid plaques. Total microglia counts are unchanged with regards to area density and the total number of microglia co-localizing to plaques is unchanged.
- FIG.14C shows Iba1+ volume
- FIG.14D shows the co-localized Iba1+ and CD68+ (as a percentage of Iba1+ staining)
- FIG.14E shows the co-localized Iba1+ and CD68+.
- Increased CD68 in microglia around plaques indicates increased phagocytic activation.
- F. Gene Expression To identify changes in gene expression following administration of lemborexant or doxepin to PSAPP mice, qPCR array was performed on cortical tissue from the mice.
- FIG.15 is a quantitative plot of three transcripts which showed significant differences in expression. Ifnb1 encodes the inflammatory mediator IFN- beta, which is implicated in microglia regulation in AD.
- Rab5a encodes a lysosomal protein
- Mmp-2 encodes a metalloprotease which has been shown to degrade A ⁇ .
- Data is displayed as fold change (relative to the mean for VEH). Error bars indicate mean ⁇ SEM, each dot is a single mouse. P values from 1-way ANOVA are shown. [0372] The results show that genes associated with A ⁇ degradation are upregulated in PSAPP mice after administration of lemborexant. The gene expression changes were not significant in the doxepin-treated group. Attorney Docket No.: 08061.0057-00304 G.
- FIG.16A shows a schematic of the experimental design (Lau et al., STAR Protoc.2021). Briefly, 5 month old PSAPP mice were treated daily with vehicle (Veh) or Lemborexant (LEM) 30mg/kg by oral gavage for 7 days. After treatment the 7th day, amyloid plaques were labeled in vivo via intraperitoneal (i.p.) injection of methoxy-X04 (MX04). Three hours later, mice were sacrificed and microglial were isolated from the brain and subjected to flow cytometry.
- Veh vehicle
- LAM Lemborexant
- FIG.16B show a flow cytometry gating strategy to isolate likely microglia. Following side and forward scatter gating to identify viable single cells, the CD45- low,CD11b+ population was isolated as likely microglia.
- FIG.16C shows the analysis of this cell population for methoxy-X04 positivity.
- FIG.17A shows a schematic of the experimental design. Briefly, 9- month-old PSAPP mice with amyloid plaques were treated daily with vehicle (Veh), or Lemborexant (LEM, 30mg/kg)) for 30 days.
- FIG.17B shows representative images of MX04, thiazine Red, and overlay images.
- FIG.17D shows a graph where at least 10 plaques were analyzed per mouse and the average per mouse is shown with each circle on the graph.
- FIG.17C shows representative images of amyloid plaques labeled with X34, microglia labeled with IBA1, and microglial phagosome labeled with CD68.
- the IBA1-CD68 colocalized volume was calculated within a 20 ⁇ m sphere from each amyloid plaque to determine peri-plaque microglial CD68 expression.
- FIG.17E shows quantification of colocalized IBA1-CD68, displayed as a percent of total IBA1 (total microglial) area. Each circle represents the average for a single mouse, with 10 plaques quantified per mouse.
- FIG.17D and FIG.17E the comparison between VEH and LEM treated groups was analyzed by two tailed t-test.
- the effects of administering doxepin were also assessed in aged PSAPP mice. Neither lemborexant nor doxepin administration led to a significant change in total plaque number in aged mice. A similar lack of significant change was observed in plaque volume, IBA1+ cells, IBA1 volume, and co-localization of IBA1- CD68.
- dystrophic neurite volume surrounding the amyloid plaques was quantified by measuring BACE1 elevation in pre-synaptic termini.
- APP/PS1 are double transgenic mice expressing a chimeric mouse/human amyloid precursor protein (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-dE9), both directed to CNS neurons.
- each cage was furnished with 160 g corncob bedding, 220 g food pellets, and 380 mL water, so that each cage weighed the same except for a small variability between mouse body weight.
- the sleep-wake states were analyzed by SleepStats software (Signal Solutions).30-second epochs were used for scoring sleep bout lengths following the manufacturer’s default setting in the current version of SleepStats software.
- mice were perfused at 4.5-mo and 5-mo, respectively. Brains were extracted and processed for IHC/IF, transcriptomics, or proteomics analyses. M. Methods to assess the effects of lemborexant and doxepin on microglial A ⁇ -phagocytic activity [0384] 5-mo PSAPP mice were dosed with VEH, or LEM (30 mg/kg/day) at ZT0 each day for 7 days. On the 7th day, mice were injected i.p.
- mice were perfused at 10-mo and brains were extracted and processed for IHC/IF analyses. Fixed brain sections were stained with thiazine red and compared to MX-04 staining to estimate plaque growth.
- O. Drugs [0386] Mice were dosed per oral with either vehicle (VEH), Doxepin (DOX, Cayman Chemical #15888, solubilized in PBS), or Lemborexant (LEM, suspended in 0.5% methylcellulose) at ZT0 (lights on). VEH mice were given similar volumes of vehicle each day. The tip of the 22-gauge oral gavage needle was dipped in 100% sucrose solution immediately prior to gavaging. MethoxyX-04 (Tocris #4920, 10 mg/kg) was injected i.p.
- mice were perfused between ZT5 and ZT7 (1100 and 1300 hours). Mice were deeply anesthetized with i.p. pentobarbital (150mg/kg), then perfused transcardially with ice-cold Dulbecco’s modified PBS (DPBS) containing 3 g/l heparin. The brains were carefully extracted and the left hemisphere was post-fixed in 4% paraformaldehyde for 48 hours (4°C), then cryoprotected with 30% sucrose in PBS (4°C) for 24 hours.
- DPBS ice-cold Dulbecco’s modified PBS
- Sections were washed again in TBS x 3, then blocked in 3% milk diluted in TBS+0.25% Triton X- 100 for 30 minutes. Sections were incubated overnight in biotinylated HJ3.4 in TBS + 0.25% Triton X-100 + 1% milk at 4°C. The next day, sections were washed and then developed using ABC Elite (Vector PK-6100) for 60 minutes. Sections were then incubated in 3,3-diaminobenzidine (DAB, Sigma-Aldrich) as chromogen and 0.05% hydrogen peroxide as substrate, and dehydrated before coverslipping using Cytoseal 60 (8310; Thermo Fisher Scientific).
- DAB 3,3-diaminobenzidine
- Quantification of confocal images for IBA1 and CD68 volume around X34+ plaques was performed on a semiautomated platform using MATLAB and Imaris 10.0.1 software (Bitplane). To create surfaces of each stain based on a threshold applied to all images, X34+ surfaces were dilated by 20 ⁇ m and colocalized with various immunostained surfaces. IBA1+ and CD68+ surface areas were then colocalized within the 20 ⁇ m extended shell around the plaques. For quantification of the number of plaque-associated IBA1+ microglia, a threshold was applied across all images to assign spots to each cell body or punctum.
- X34 surfaces were dilated to 20 ⁇ m, and spots were counted within the X34+ dilated Attorney Docket No.: 08061.0057-00304 surface. Any spots fully within or partially touching the extended surface were included in the analysis.
- R. Western blotting [0392] Tissue samples were homogenized by sonication on ice in radioimmunoprecipitation (RIPA) buffer (Pierce, Thermo Scientific) containing complete protease inhibitors and PhosSTOP phosphatase inhibitors (Roche). PAGE and Western blotting were performed using Invitrogen Novex gels and reagents.
- RIPA radioimmunoprecipitation
- Tukey for equal group sizes
- Tukey-Kramer for unequal group sizes
- an F test was first performed for datasets with a single dependent variable and 2 groups, to determine if variances were significantly different. If not, a 2-tailed unpaired t-test was performed. If variances were different, a non-parametric Mann- Whitney U test was performed. Outliers were identified using the Grubbs test and were excluded. All P values are noted in the figures.
- Control mice or Bmal1 KO mice were treated with tamoxifen to delete Bmal1.
- One month later infra-red actigraphy was recorded under 12h:12h L:D condition (yellow area) or constant darkness (DD condition).
- Mice were administered lemborexant (LEM) at a dose of 30mg/kg by oral gavage at 6am (previous ZT 0) each day for 9 days (indicated by red bar and red arrow). Actigraphy was collected for 2 more weeks after dosing ceased.
- FIG.18A shows representative actograms for control and Bmal1 knockout mice.
- FIG.18B is a quantification of actigraphic endpoints during different portions of the experiment (in FIG.18A, LD indicates during yellow area, DD+LEM is area with the red bar, and DD is the rest of the recording). Data analyzed by 2-way ANOVA with Tukey post-hoc test. [0396] The results indicate that Bmal1 knockout mice showed greater daytime activity than wild-type mice under all conditions (LD, DD + LEM, and DD). Bmal1 KO mice are able to maintain their locomotor activity in DD conditions similar to LD when they are given lemborexant daily at CT0 (CT0 is similar to ZT0 in DD conditions).
- Relative amplitudes which indicate the ratio of average activity in the most active 10 hours as compared to average activity in the least active 5 hours, were lower in Bmal1 knockout mice under all conditions.
- higher relative Attorney Docket No.: 08061.0057-00304 amplitudes correlate with stable rhythms.
- Interdaily stability (IS) which measures the synchronization between daily 24-hour rhythms, was lower in Bmal1 knockout mice under all conditions. High IS indicates good synchronization of rhythms.
- IS Interdaily stability
- compositions of the disclosure e.g., any, composition, therapeutic or active ingredient; any method of production; any method of use; etc.
- any particular embodiment of the compositions of the disclosure can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Urology & Nephrology (AREA)
- Hematology (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Neurology (AREA)
- Cell Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Food Science & Technology (AREA)
- Neurosurgery (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
Abstract
Description
Claims
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2023347307A AU2023347307A1 (en) | 2022-09-23 | 2023-09-22 | Methods of reducing neurodegeneration associated with neurodegenerative diseases |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263376949P | 2022-09-23 | 2022-09-23 | |
US63/376,949 | 2022-09-23 | ||
US202263382278P | 2022-11-03 | 2022-11-03 | |
US63/382,278 | 2022-11-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024064897A1 true WO2024064897A1 (en) | 2024-03-28 |
Family
ID=88506615
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2023/074913 WO2024064897A1 (en) | 2022-09-23 | 2023-09-22 | Methods of reducing neurodegeneration associated with neurodegenerative diseases |
Country Status (3)
Country | Link |
---|---|
AU (1) | AU2023347307A1 (en) |
TW (1) | TW202425995A (en) |
WO (1) | WO2024064897A1 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11026944B2 (en) | 2014-10-23 | 2021-06-08 | Eisai R&D Management Co., Ltd. | Compositions and methods for treating insomnia |
US11096941B2 (en) | 2016-05-12 | 2021-08-24 | Eisai R&D Management Co.. Ltd. | Methods of treating circadian rhythm sleep disorders |
WO2023283650A1 (en) | 2021-07-09 | 2023-01-12 | Eisai R&D Management Co., Ltd. | Biomarkers for alzheimer's disease treatment |
-
2023
- 2023-09-22 TW TW112136326A patent/TW202425995A/en unknown
- 2023-09-22 WO PCT/US2023/074913 patent/WO2024064897A1/en active Application Filing
- 2023-09-22 AU AU2023347307A patent/AU2023347307A1/en active Pending
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11026944B2 (en) | 2014-10-23 | 2021-06-08 | Eisai R&D Management Co., Ltd. | Compositions and methods for treating insomnia |
US11096941B2 (en) | 2016-05-12 | 2021-08-24 | Eisai R&D Management Co.. Ltd. | Methods of treating circadian rhythm sleep disorders |
EP4056180A1 (en) * | 2016-05-12 | 2022-09-14 | Eisai R&D Management Co., Ltd. | Methods of treating circadian rhythm sleep disorders |
WO2023283650A1 (en) | 2021-07-09 | 2023-01-12 | Eisai R&D Management Co., Ltd. | Biomarkers for alzheimer's disease treatment |
Non-Patent Citations (21)
Title |
---|
"Alzheimer's Association, Alzheimer's Association report, 2010 Alzheimer's disease facts and figures", ALZHEIMER DEMENT, vol. 6, 2010, pages 158 - 94 |
ALSHAER H ET AL.: "Reproducibility and predictors of the apnea hypopnea index across multiple nights", SLEEP SCI., vol. 11, no. 1, 2018, pages 28 - 33 |
BERG, L. ET AL.: "Mild senile dementia of the Alzheimer type: 2. Longitudinal assessment", ARIN. NEUROL., vol. 23, 1988, pages 477 - 84 |
BROOKMEYER, R. ET AL.: "Forecasting the global burden of Alzheimer's Disease", ALZHEIMER DEMENT, vol. 3, 2007, pages 186 - 91, XP022100576, DOI: 10.1016/j.jalz.2007.04.381 |
BUYSSE D. J. ET AL.: "The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research", PSYCHIATRY RES., vol. 28, no. 2, 1989, pages 193 - 213, XP001020700, DOI: 10.1016/0165-1781(89)90047-4 |
CHUNG F. ET AL.: "STOP-Bang Questionnaire: A Practical Approach to Screen for Obstructive Sleep Apnea", CHEST, vol. 149, no. 3, 2016, pages 631 - 638 |
DRZEZGA, A. ET AL.: "Effect of APOE genotype on amyloid plaque load and gray matter volume in Alzheimer disease", NEUROLOGY, vol. 72, 2009, pages 1487 - 94 |
ENGLUND, H. ET AL., J. NEUROCHEM., vol. 103, 2007, pages 334 - 45 |
FOLSTEIN, M.F. ET AL.: "Mini-mental state. A practical method for grading the cognitive state of subjects for the clinician", J. PSYCHIATR. RES., vol. 12, 1975, pages 189 - 98 |
GAO FAN ET AL: "The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target", NEUROSCIENCE LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 765, 14 September 2021 (2021-09-14), XP086847164, ISSN: 0304-3940, [retrieved on 20210914], DOI: 10.1016/J.NEULET.2021.136247 * |
HEBERT, L.E. ET AL.: "Alzheimer disease in the U.S. population: prevalence estimates using the 2000 census", ARCH NEUROL, vol. 60, 2003, pages 1119 - 1122 |
KIRMESS ET AL., J. CIINICA CHIMICA ACTA, vol. 519, 2021, pages 267 - 275 |
KIRMESS ET AL., J. CLINICA CHIMICA ACTA, vol. 519, 2021, pages 267 - 275 |
KLUNK WE ET AL.: "The Centiloid Project: standardizing quantitative amyloid plaque estimation by PET", ALZHEIMER'S DEMENT, vol. 11, 2015, pages 1 - 15 |
KLUNK WE ET AL.: "The Centiloid Project: standardizing quantitative amyloid plaque estimation by PET", ALZHEIMER'S DEMENT., vol. 11, 2015, pages 1 - 15 |
LAU ET AL., STAR PROTOC, 2021 |
MCKHANN, G. M. ET AL.: "The diagnosis of dementia due to Alzheimer's disease: Recommendations from the National Institute on Aging—Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease", ALZHEIMER DEMENT, vol. 7, 2011, pages 263 - 9, XP028243208, DOI: 10.1016/j.jalz.2011.03.005 |
MCKHANN, G.M. ET AL.: "The diagnosis of dementia due to Alzheimer's disease: Recommendations from the National Institute on Aging - Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease", ALZHEIMER DEMENT., vol. 7, 2011, pages 263 - 9, XP028243208, DOI: 10.1016/j.jalz.2011.03.005 |
ROSEN, W. G. ET AL.: "A new rating scale for Alzheimer's disease", AM. J. PSYCHIATRY, vol. 141, 1984, pages 1356 - 64 |
WANG, J. ET AL.: "ADCOMS: a composite clinical outcome for prodromal Alzheimer's disease trials", J. NEUROL. NEUROSURG. PSYCHIATRY, vol. 87, 2016, pages 993 - 999 |
ZHOU FANG ET AL: "Suvorexant ameliorates cognitive impairments and pathology in APP/PS1 transgenic mice", NEUROBIOLOGY OF AGING, vol. 91, 1 July 2020 (2020-07-01), US, pages 66 - 75, XP093116722, ISSN: 0197-4580, Retrieved from the Internet <URL:https://pdf.sciencedirectassets.com/271067/1-s2.0-S0197458020X00066/1-s2.0-S0197458020300531/main.pdf?X-Amz-Security-Token=IQoJb3JpZ2luX2VjED4aCXVzLWVhc3QtMSJHMEUCIQDP6qlVNFdFeyE0Dc98UwbJX9d0RL7QCE3b3WyoXyZRrAIgSjFcrJIKUfrXC+nP9IvF634MnqSsy2QWHUJxffUxBGYquwUI1///////////ARAFGgwwNTkwMDM1NDY4NjUiDMdj4> DOI: 10.1016/j.neurobiolaging.2020.02.020 * |
Also Published As
Publication number | Publication date |
---|---|
AU2023347307A1 (en) | 2025-03-13 |
TW202425995A (en) | 2024-07-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Gao et al. | Tau in Alzheimer's disease: mechanisms and therapeutic strategies | |
Das et al. | A close look at BACE1 inhibitors for Alzheimer’s disease treatment | |
Parihar et al. | Mitoenergetic failure in Alzheimer disease | |
CA3079259A1 (en) | Ganaxolone for use in treating genetic epileptic disorders | |
US20200338040A1 (en) | Methods for treating alzheimer's disease and related disorders | |
Ortiz‐González et al. | Homozygous boricua TBCK mutation causes neurodegeneration and aberrant autophagy | |
US20230078820A1 (en) | Fenfluramine for treatment of demyelinating diseases and conditions | |
US20190298740A1 (en) | Methods and compositions for treating hallucinations and conditions related to the same | |
US9388413B2 (en) | Method for selectively inhibiting ACAT1 in the treatment of neurodegenerative diseases | |
Li et al. | Ube2c-inhibition alleviated amyloid pathology and memory deficits in APP/PS1 mice model of AD | |
JP6353110B2 (en) | Tau aggregation inhibitor | |
KR20190102181A (en) | A composition comprising an anti-Abeta primitive fibrous antibody and a beta-secretase BACE1 inhibitor for the treatment of Alzheimer's disease | |
US10328051B2 (en) | Proline or proline derivatives for the treatment of dementia | |
JP4971794B2 (en) | Methods for reducing amyloid deposition, amyloid neurotoxicity and microgliosis | |
US20220105106A1 (en) | Compositions and methods relating to use of agonists of alpha5-containing gabaa receptors | |
US10100308B2 (en) | Method for selectively inhibiting ACAT1 in the treatment of neurodegenerative diseases | |
AU2023347307A1 (en) | Methods of reducing neurodegeneration associated with neurodegenerative diseases | |
US9388414B2 (en) | Method for selectively inhibiting ACAT1 in the treatment of neurodegenerative diseases | |
US20210113552A1 (en) | Methods for enhancing cellular clearance of pathological molecules via activation of the cellular protein ykt6 | |
Wu et al. | Intermittent Fasting Ameliorates β‐Amyloid Deposition and Cognitive Impairment Accompanied by Decreased Lipid Droplet Aggregation Within Microglia in an Alzheimer's Disease Model | |
US20240189307A1 (en) | Methods of stabilizing the neuronal proteome against collapse and protecting vascular cells | |
Ferreira | Establishing the relevance of Tau isoform imbalance in the onset and progression of Machado-Joseph disease | |
Kamal et al. | Neuroscientific Research for Management of Dementia | |
Ali et al. | Molecular mechanisms and biomarkers in neurodegenerative disorders: a comprehensive review | |
WO2023230560A1 (en) | Treatment of organic acidemias or pantothenate kinase associated neurodegeneration with modulators of pantothenate kinases |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23793634 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: AU2023347307 Country of ref document: AU |
|
ENP | Entry into the national phase |
Ref document number: 2023347307 Country of ref document: AU Date of ref document: 20230922 Kind code of ref document: A |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112025005466 Country of ref document: BR |