WO2022251048A1 - Anti-amyloid beta antibodies and uses thereof - Google Patents

Anti-amyloid beta antibodies and uses thereof Download PDF

Info

Publication number
WO2022251048A1
WO2022251048A1 PCT/US2022/030167 US2022030167W WO2022251048A1 WO 2022251048 A1 WO2022251048 A1 WO 2022251048A1 US 2022030167 W US2022030167 W US 2022030167W WO 2022251048 A1 WO2022251048 A1 WO 2022251048A1
Authority
WO
WIPO (PCT)
Prior art keywords
human subject
weeks
apoe
brain
antibody
Prior art date
Application number
PCT/US2022/030167
Other languages
French (fr)
Inventor
John Randall II SIMS
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Publication of WO2022251048A1 publication Critical patent/WO2022251048A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present disclosure is related to methods of preventing or treating a disease with anti-amyloid beta antibodies, wherein the disease is characterized by deposition of amyloid beta (Ab) in a human subject.
  • the present disclosure is also related to doses and dosing regimens of the anti-Ab antibodies useful for treating or preventing a disease characterized by deposition of Ab.
  • Some aspects of the present disclosure are related to treating or preventing a disease characterized by deposition of Ab in human subjects, wherein the human subjects are selected, and/or treated with the antibodies of the present disclosure, based on the presence of one or two alleles of APOE e4 in the subject’s genome.
  • the diseases that can be treated or prevented using antibodies, dosing regimens, or methods disclosed herein include, e.g ., Alzheimer’s disease (AD), Down’s syndrome, and cerebral amyloid angiopathy (CAA).
  • AD Alzheimer’s disease
  • CAA cerebral amyloid angiopathy
  • the present disclosure is related to slowing cognitive decline, functional decline, and/or memory loss in the subject.
  • the present disclosure is related to reducing amyloid load in the brain of the human subject.
  • AD Alzheimer’ s disease
  • AD Alzheimer’ s disease
  • cognitive and functional impairment Selkoe, “The Origins of Alzheimer Disease: A is for Amyloid,” JAMA 283:1615-7 (2000); Hardy etal. , “The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics,” Science 297:353-6 (2002); Masters etal. , “Alzheimer’s Disease,” Nat. Rev. Dis. Primers 1:15056 (2015); and Selkoe et al, “The Amyloid Hypothesis of Alzheimer’s Disease at 25 years,” EMBO Mol. Med. 8:595-608 (2016)).
  • Amyloid beta is formed by the proteolytic cleavage of a larger glycoprotein called amyloid precursor protein (APP).
  • APP is an integral membrane protein expressed in many tissues, but especially in neuron synapses.
  • APP is cleaved by g-secretase to release the Ab peptide, which encompasses a group of peptides ranging in size from 37-49 amino acid residues.
  • Ab monomers aggregate into several types of higher order structures including oligomers, protofibrils, and amyloid fibrils.
  • Amyloid oligomers are soluble and may spread throughout the brain, while amyloid fibrils are larger, insoluble, and can further aggregate to form amyloid deposits or plaques.
  • amyloid deposits found in human patients include a heterogeneous mixture of Ab peptides, some of which include N-terminal truncations and further may include N-terminal modifications such as an N-terminal pyroglutamate residue (pGlu).
  • pGlu N-terminal pyroglutamate residue
  • amyloid deposits in driving disease progression is supported by study of uncommon genetic variants that either increase or decrease Ab deposition (Fleisher et al ., “Associations Between Biomarkers and Age in the Presenilin 1 E280A Autosomal Dominant Alzheimer Disease Kindred: A Cross-sectional Study,” JAMA Neurol 72:316-24 (2015); Jonsson etal. , “A Mutation in APP Protects Against Alzheimer’s Disease and Age- related Cognitive Decline,” Nature 488:96-9 (2012)).
  • Antibodies targeting amyloid are known, including, e.g., bapineuzumab, gantenerumab, aducanumab, GSK933776, solanezumab, crenezumab, ponezumab, and lecanemab (BAN2401).
  • Some antibodies targeting amyloid have shown promise as a therapeutic for Alzheimer’s disease in preclinical studies. Despite this promise, most antibodies targeting amyloid have failed to meet therapeutic endpoints in multiple clinical trials. For example, solanezumab, an anti-Ab, has been studied in multiple Phase 3 clinical trials but did not meet its clinical endpoint. See Honig, et al.
  • Ab antibodies have led to adverse events in humans, such as, amyloid-related imaging abnormalities (ARIA), suggestive of vasogenic edema and sulcal effusions (ARIA-E), microhemorrhages and hemosiderin deposits (ARIA-H), infusion site reactions, and risk of immunogenicity.
  • ARIA amyloid-related imaging abnormalities
  • ARIA-E vasogenic edema and sulcal effusions
  • ARIA-H microhemorrhages and hemosiderin deposits
  • infusion site reactions and risk of immunogenicity.
  • some antibody treatment programs implement dose-titration schemes that include multiple dose escalations (3-4 steps) over a period of about 6-months prior to reaching their efficacious dose level. See , e.g, Budd et al ., “Clinical Development of Aducanumab, an Anti-Ab Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer's Disease,” The Journal of Prevention of Alzheimer's Disease 4.4:255 (2017) and Klein et al., “Gantenerumab Reduces Amyloid-b Plaques in Patients with Prodromal to Moderate Alzheimer’s Disease: a PET Substudy Interim Analysis,” Alzheimer's Research & Therapy 11.1:101 (2019). Such treatment regimens may not fully clear amyloid plaques or may delay clearance of amyloid plaque.
  • Alzheimer’s disease is still principally diagnosed and treated based on symptoms, e.g. , like a psychiatric illness, rather than based on brain pathology.
  • This causes clinical trials to include heterogenous populations (e.g, with wide variation in levels of underlying pathology and/or different underlying diseases) which makes replicating clinical trial data very challenging.
  • determining whether subjects having Ab deposits may respond to an anti-Ab antibody treatment is uniquely challenging and the task of properly identifying whether a patient may respond to anti-Ab antibody treatments is of utmost importance for, e.g, a timely referral to a memory clinic, a correct and early AD diagnosis, initiation of symptomatic treatment, future planning, and initiating disease-modifying treatments.
  • Fig. 1 shows a table describing the effect size for APOE e4 carrier vs. non-carrier across Expedition-3 and Trailblazer-1 Clinical Trials.
  • iADRS Integrated Alzheimer's Disease (AD) Rating Scale;
  • ADAS-Cog Alzheimer's Disease Assessment Scale cognitive subscale;
  • iADL instrumental Activities of Daily Living;
  • CDR-SB Clinical Dementia Rating scale - sum of boxes;
  • MMSE Mini-Mental State Exam;
  • ADAS Alzheimer's Disease Assessment Scale;
  • FAQ Function Activities Questions.
  • Fig. 2 shows the effect size for APOE e4 carrier vs. non-carrier. Effect sizes of Mild AD in Sola (not amyloid selected).
  • Fig. 3 shows change in cognition ADAS-Cogl4 (primary endpoint) from the Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS- Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 4A shows change in cognition ADAS-Cogl4 by APOE e4 status from the Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, patients that are carriers of APOE e4. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error
  • APOE Apolipoprotein E.
  • Fig. 4B shows change in cognition ADAS-Cogl4 by APOE e4 status from Expedition- 3 Clinical Trial of Solanezumab initiated in patients with mild AD dementia, patients that are not carriers of APOE e4. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error
  • APOE Apolipoprotein E.
  • AD Alzheimer’s disease
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS latitude squares
  • n number
  • SE Standard Error
  • APOE apolipoprotein E.
  • Fig. 5B shows change in cognition ADAS-Cogl4 by APOE e4 status.
  • This data related to pooled Expedition- 1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 non-carriers. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error
  • APOE apolipoprotein E.
  • Fig. 6 shows change in complex ADLs - ADCS-iADL in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADL Activities of Daily Living
  • ADCS- iADL AD Cooperative Study -Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 7A shows change in ADLs - ADCS-iADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers.
  • Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • Fig. 7B shows change in ADLs - ADCS-iADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non carriers.
  • Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • ADCS- iADL AD Cooperative Study -Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • Fig. 8A shows change in ADCS-iADLs by APOE e4 status in pooled Expedition-1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADCS- iADL AD Cooperative Study -Instrumental Activities of Daily Living
  • LS least squares
  • SE Standard Error
  • n number.
  • Fig. 8B shows change in ADCS-iADLs by APOE e4 status in pooled Expedition- 1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 non carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • ADCS- iADL AD Cooperative Study -Instrumental Activities of Daily Living
  • LS least squares
  • SE Standard Error
  • n number.
  • Fig. 9 shows change in cognition-MMSE in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • LS least squares
  • MMSE Mini-Mental State Examination
  • n number
  • SE Standard Error.
  • Fig. 10A shows change in cognition-MMSE by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers.
  • the APOE s4 carrier status-by- treatment interaction displayed a p-value of 0.201. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • MMSE Mini-Mental State Examination
  • LS least squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • Fig. 10B shows change in cognition-MMSE by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non-carriers.
  • the APOE e4 carrier status-by-treatment interaction displayed a p-value of 0.201. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • MMSE Mini-Mental State Examination
  • LS least squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • Fig. 11 shows change in composite scale CDR-SB in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • CDR-SB Clinical Dementia Rating Sum of Boxes
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 12A shows change in composite scale CDR-SB by APOE e4 status in Expedition- 3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • CDR- SB Clinical Dementia Rating Sum of Boxes
  • LS least squares
  • n number
  • SE Standard Error
  • APOE/Apoe apolipoprotein.
  • Fig. 12B shows change in composite scale CDR-SB by APOE e4 status in Expedition- 3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • CDR- SB Clinical Dementia Rating Sum of Boxes
  • LS least squares
  • n number
  • SE Standard Error
  • APOE/Apoe apolipoprotein.
  • Fig. 13 shows change in ADCS-ADL in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADL Activities of Daily Living
  • ADCS-ADL AD Cooperative Study-Activities of Daily Living
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 14A shows change in ADCS-ADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers.
  • the APOE s4 carrier status-by-treatment interaction displayed a p-value of 0.714. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • AD Alzheimer’ s disease
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS latitude squares
  • n number
  • APOE apolipoprotein E
  • SE Standard Error.
  • Fig. 15 shows change in composite scale: iADRS in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • iADRS Integrated AD Rating Scale
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 16 shows change in cognition and ADLs at 80 weeks in Expedition-3 clinical trial of Solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • ADL Activities of Daily Living
  • ADAS- Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • ADCS-ADL AD Cooperative Study-Activities of Daily Living
  • CDR-SB Clinical Dementia Rating Sum of Boxes
  • FAQ Function Activities Questionnaire
  • iADRS Integrated AD Rating Scale
  • LS least squares
  • MMSE Mini-Mental State Examination
  • SE standard error.
  • Fig. 17 shows baseline demographics pooled from Expedition- 1 and Expedition-2 clinical trials of Solanezumab in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • Fig. 18 shows baseline clinical characteristics of Expedition-3 clinical trial of solanezumab initiated in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AChEI acetylcholinesterase inhibitor
  • ADCS-ADL AD Cooperative Study-Activities of Daily Living
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • CDR- SB Clinical Dementia Rating Sum of Boxes
  • FAQ Function Activities Questionnaire
  • MMSE Mini-Mental State Examination
  • n number
  • NA not available from topline results.
  • Fig. 19 shows baseline clinical characteristics pooled from Expedition- 1 and Expedition-2 clinical trials of solanezumab in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • AChEI acetyl cholinesterase inhibitor
  • ADCS-ADL AD Cooperative Study-Activities of Daily Living
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • CDR-SB Clinical Dementia Rating Sum of Boxes
  • FAQ Function Activities Questionnaire
  • n number
  • MMSE Mini-Mental State Examination
  • N/A not applicable
  • SD standard deviation.
  • Fig. 20A shows change in complex ADLs: ADCS-iADL from Expedition-1 clinical Trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • ADL Activities of Daily Living
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 20B shows change in complex ADLs: ADCS-iADL from Expedition-2 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADL Activities of Daily Living
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 20C shows change in complex ADLs: ADCS-iADL pooled from Expedition-1 and Expedition-2 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADL Activities of Daily Living
  • ADCS-iADL AD Cooperative Study-Instrumental Activities of Daily Living
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 20D shows change in complex ADLs: ADCS-iADL from Expedition-3 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • AD Alzheimer’s disease
  • ADAS- Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS laeast squares
  • n number
  • SE Standard Error.
  • Fig. 21B shows change in cognition: ADAS-Cogl4 from Expedition-2 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS- Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 21C shows change in cognition: ADAS-Cogl4 pooled from Expedition-1 and -2 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’s disease
  • ADAS-Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error.
  • Fig. 21D shows change in cognition: ADAS-Cogl4 pooled from Expedition-3 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study.
  • AD Alzheimer’ s disease
  • ADAS- Cogl4 AD Assessment Scale-Cognitive 14-item Subscale
  • LS least squares
  • n number
  • SE Standard Error.
  • One aspect of the present disclosure provides for doses and dosing regimens of solanezumab circumventing problematic adverse events, such as ARIA with vasogenic edema, which have been observed in patients receiving therapeutic antibodies that bind to deposited amyloid and has been dose limiting for clinical development programs. Additionally, aspects of the present disclosure provide improved methods for identifying patients for treatment of AD and other forms of dementia, CAA and/or Down’s syndrome patients with anti-amyloid beta antibodies.
  • the present disclosure involves administering doses of solanezumab or other anti-Ab antibodies to patients who have one or two alleles of APOE s4 as a means of treating patients with AD, preventing AD, and/or slowing cognitive/functional decline in patients.
  • the patients can be stratified/identified/selected/treated based on the amount of tau present in the subject’s brain (e.g, in the whole brain or in portions of the brain) and the presence of one or two alleles of APOE e4. In some embodiments, the patients can be stratified/identified/selected/treated based on the stages of AD progression (e.g. , based on the spread of tau in the brain) and the presence of one or two alleles of APOE s4.
  • Some aspects of the present disclosure provide for a dosing regimen where a human subject, suffering from a disease characterized by Ab deposits in their brain, is administered solanezumab in one step or two steps.
  • the human subject is administered one or more doses of the antibodies of the present disclosure, optionally, once about every 4 weeks.
  • the human subject is administered one or more doses of solanezumab in a first step, wherein each dose is administered once about every 4 weeks. About four weeks after administering the one or more first doses, the human subject is administered one or more second doses in a second step.
  • Some aspects of the present disclosure are related to identifying the stage/progression of AD in a patient based on i) the global or overall tau burden in the brain of a human subject or ii) the spread of tau in the subject’s brain or portions thereof.
  • the stratification of patients based on amount of tau in the brain or AD progression in portions of brain can be used to determine, e.g. , whether a patient will respond to treatments.
  • Stratification/selection of patient population based on amount of tau in the brain or AD progression in portions of brain is also helpful in solving the patient heterogeneity and replicability problems faced during design and performance of clinical trials. See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
  • the patients can be stratified/identified/selected/treated based on the amount of tau present in the subject’s brain (e.g, in the whole brain or in portions of the brain). See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
  • the patients are stratified/identified/selected/treated based on stages of AD progression (e.g, based on the spread of tau in the brain). For example, during some stages, tau burden in an AD patient is isolated to frontal lobe or regions of the temporal lobe that do not include the posterolateral temporal region (PLT).
  • PKT posterolateral temporal region
  • AD Alzheimer's disease
  • tau burden in an AD patient is limited to the posterolateral temporal (PLT) or occipital regions.
  • PLT posterolateral temporal
  • Yet another stage of AD is when the tau burden in an AD patient is present in the parietal or precuneus region or in the frontal region along with tau burden in PLT or occipital regions. See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
  • the responsive human subjects include human subjects having low to moderate tau burden or very low to moderate tau burden, optionally having one or more alleles of APOE e4.
  • the responsive human subjects exclude human subjects with high tau burden, optionally having one or more alleles of APOE s4.
  • solanezumab is administered to the responsive human subjects for treatment or prevention of a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject. See, e.g ., International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
  • Some aspects of the present disclosure are related to a method of treating or preventing a disease characterized by Ab deposits in the brain of a human subject wherein the human subject is clinically asymptomatic.
  • This method includes administering to an APOE e4 carrier (e.g, one who has one or more of the APOE e4 alleles) a dose of solanezumab.
  • an APOE e4 carrier e.g, one who has one or more of the APOE e4 alleles
  • the dose may be 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, or 1600 mg every 4 weeks. Of course, other doses may be used.
  • This may be giving an initial dose of 400 mg, and either maintaining the patient at 400 mg or titrating up to 800 mg every 4 weeks, or titrating up 1200 mg every 4 weeks, or titrating up 1600 mg over time.
  • Other embodiments may involve giving an initial dose of 1600 mg and then maintaining at that dose or titrating down to 400 mg, 800 mg, or 1200 mg.
  • Those skilled in the art will appreciate how to titrate up or down the solanezumab dose, or to maintain the patient on a particular dose (and the timing associated with making a dosing change).
  • solanezumab may be based upon numerous factors, including PET scans, clinical observations, performance by the patient on various “tests,” etc.
  • the clinically asymptomatic subjects are known to have an Alzheimer's disease-causing genetic mutation.
  • “clinically asymptomatic subjects known to have an Alzheimer's disease-causing genetic mutation” include patients known to have a PSEN1 E280A Alzheimer's disease-causing genetic mutation (Paisa mutation), a genetic mutation that causes autosomal-dominant Alzheimer's disease or are at higher risk for developing AD by virtue of carrying one or two APOE e4 alleles.
  • anti-Ab antibody refers to an antibody that binds to an epitope present on Ab. In some embodiments, the anti-Ab antibody binds to a soluble form of Ab. In other embodiments, the anti-Ab antibody binds to an insoluble form of Ab, such as Ab plaques. In some embodiments, the anti-Ab antibody binds an epitope present in Ab1-40 or Ab ⁇ - 42. In other embodiments, the anti-Ab antibody binds an epitope present in a truncated form of Ab1-40 or Ab1-42, for example, a truncated form lacking 1-20 N terminal amino acids and/or lacking 1-20 C-terminal amino acids.
  • the anti-Ab antibody binds an epitope present in a fragment of Ab1-40 or Ab1-42 and having a length of about 5-20 amino acids.
  • Anti-Ab antibodies have been disclosed in the art. (See, e.g., U S. Patent Nos.
  • One aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having an APOE e4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subj ect that has one or two alleles of APOE e4.
  • the antibody administered to the human subject is an anti-Ab antibody.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
  • the antibody administered to the human subject is an anti-Ab antibody.
  • Yet another aspect of the present disclosure is related to method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE s4 allele; and ii) if the human subject is identified as having an APOE e4 allele, administering or having administered a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
  • the antibody administered to the human subject is an anti-Ab antibody.
  • the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE s4 allele.
  • the antibody administered to the human subject is an anti-Ab antibody.
  • One aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject having an APOE e4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subject that has one or two alleles of APOE e4.
  • Another aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
  • An aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE e4 allele; and ii) if the human subject is identified as having an APOE s4 allele, administering or having administered an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
  • Another aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE s4 allele.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising administering to the human subject an effective amount of solanezumab, wherein the human subject has been determined as having a very low to moderate tau burden or low to moderate tau burden.
  • the human subject has one or two alleles of APOE e4.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have one or two alleles of APOE e4 and very low to moderate tau burden or low to moderate tau burden comprising: administering to the human subject an effective amount of solanezumab.
  • the human subject has one or two alleles of APOE e4.
  • the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined as not having a high tau burden comprising: administering to the human subject an effective amount of solanezumab.
  • the human subject has one or two alleles of APOE e4.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has low to moderate tau burden or a very low to moderate tau burden; and if the human subject has low to moderate tau burden or a very low to moderate tau burden, then: administering to the human subject an effective amount of solanezumab.
  • the human subject has one or two alleles of APOE e4.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising administering to the human subject an effective amount of solanezumab, wherein the human subject has been determined as not having a high tau burden.
  • the human subject has one or two alleles of APOE e4.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has high tau burden; and if the human subject does not have high tau burden, then: administering to the human subject an effective amount of solanezumab.
  • the human subject has one or two alleles of APOE e4.
  • an anti-Ab antibody may be used to decrease, prevent further increase of tau burden, or slow the rate of tau accumulation in different portions of a human brain, e.g ., in different lobes of the brain of a human subject.
  • solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the frontal lobe of the human brain.
  • solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the parietal lobe of the human brain.
  • solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the occipital lobe of the human brain.
  • solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the temporal lobe of the human brain. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the posterolateral temporal lobe.
  • An aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the temporal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the temporal lobe of the brain and administering an anti-Ab to the human subject.
  • the human subject has tau burden in the posterolateral temporal lobe.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the occipital lobe of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the occipital lobe of the brain and administering an anti-Ab to the human subject.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the parietal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the parietal lobe of the brain and administering an anti-Ab to the human subject.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in the frontal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the frontal lobe of the brain and administering an anti-Ab to the human subject.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in the posterolateral temporal (PLT) and/or occipital lobe of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the posterolateral temporal (PLT) and/or occipital lobe of the brain and administering an anti-Ab to the human subject.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in i) parietal or precuneus region or ii) in frontal region along with tau burden in PLT or occipital regions of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits comprising determining whether the human subject has tau burden in i) parietal or precuneus region or ii) in the frontal region along with tau burden in PLT or occipital regions of the brain and administering an anti-Ab to the human subject.
  • Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain wherein the method comprises administering an anti-Ab to the human subject.
  • Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits comprising determining whether the human subject has tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain and administering an anti-Ab to the human subject.
  • PHT posterolateral temporal region
  • the present disclosure is related to a method of selecting a human subject for treatment or prevention of a disease characterized by amyloid beta deposits in the brain of a human subject.
  • the human subject is selected based on the amount of global (overall) tau in the brain of the human subject.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has very low to moderate tau in the brain.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has low to moderate tau (or intermediate tau) in the brain.
  • the human subject is excluded from treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has high tau in the brain.
  • the human subject is selected based on progression of AD in the brain of the human subject. For example, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the frontal lobe of the brain. In another embodiment, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the parietal lobe of the brain.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the occipital lobe of the brain.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the temporal lobe of the brain.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the posterolateral temporal (PLT) and/or occipital lobe of the brain.
  • PHT posterolateral temporal
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in i) parietal or precuneus region or ii) in frontal region along with tau burden in PLT or occipital regions of the brain.
  • the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain.
  • PLT posterolateral temporal region
  • the subject described in the various aspects of the present disclosure has been determined to have a posterior-lateral temporal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe and occipital lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, and parietal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe, and frontal lobe tau burden.
  • the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe and / or frontal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe and / or frontal lobe tau burden corresponds a neurological tau burden of greater than 1.46 SUVr based on PET imaging. In some embodiments, tau burden in a portion of the human brain ( e.g ., in a lobe of the brain) can be used to determine whether administration of the anti-Ab should be discontinued.
  • solanezumab is administered to the subject until there is a decrease, prevention of further increase, or slowing in the rate of tau burden/accumulation in the temporal lobe, the occipital lobe, the parietal lobe, or the frontal lobe.
  • the tau burden present in a portion of the brain of a human subject can be used for selection of optimal treatment regimens or for administration of therapeutic modalities in combination with solanezumab.
  • the presence of tau burden in the frontal lobe of the brain of an amyloid positive human subject can be used as a metric to determine whether the human subject will benefit from administration of an anti-Ab antibody alone or its combination with an anti-tau antibody.
  • solanezumab in combination with an anti-tau antibody may be used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in different portions of a human brain, e.g., in different lobes of the brain of a human subject.
  • the tau burden in different portions of a human brain e.g, in different lobes of the brain of a human subject can be used for i) tracking patient’s response to treatment, or ii) when a therapy may need to be reinitiated.
  • the antibodies, methods, or dosing regimens described in various aspects of the present disclosure cause: i) reduction in Ab deposits in the brain of the human subject and/or ii) slow cognitive decline or functional decline in the human subject. In some embodiments, the antibodies, methods, or dosing regimens described herein results in reduction of amyloid plaques.
  • Table 1 below shows how administration of solanezumab affects the amount of tau in various lobes of the brain, and how that amount differs based upon whether the patient is an APOE e4 carrier or non-carrier.
  • solanezumab is administered to the subject for a duration sufficient to treat or prevent the disease. In some embodiments, solanezumab (including the first doses of the antibody and the second doses of the antibody) is administered to the subject for a duration of up to about 54 weeks, about 72 weeks, or about 80 weeks, optionally, about once every 4 weeks or about once every month.
  • the antibody of the present disclosure is administered to the patient till amyloid levels in the brain of the patient reach a normal range. In some embodiments, the antibody of the present disclosure is administered to the subject till amyloid deposits or plaques are cleared from the subj ecf s brain. In some embodiments, the antibody is administered to the subject till the subject reaches amyloid negative status (defined as ⁇ 24.1 CL amyloid plaque).
  • normal range of amyloid plaque in brain is used interchangeably with brain amyloid plaque is “cleared.”
  • the normal range of amyloid plaque is defined as demonstrating an amyloid plaque level of 25 centiloids or lower for two consecutive PET scans at least 6 months apart or a single PET scan demonstrating a plaque level of less than 11 centiloids.
  • the dose of solanezumab is every 4 weeks (possibly modifying the dose every 4 weeks, as outlined herein) for up to about 54 weeks, about 72 weeks, or about 80 weeks.
  • solanezumab slows disease progression in patients with early symptomatic Alzheimer’s disease (AD) and with the presence of intermediate brain tau burden.
  • the antibody of the present disclosure is administered to the subject as one dose or more than one doses.
  • the patients receive solanezumab at a dose of 400 mg given intravenously every 4 weeks for 76 weeks or 80 weeks or some other time period.
  • the dose of solanezumab may be increased to 800 mg, 1200 mg, or 1600 mg given every 4 weeks.
  • the antibody of the present disclosure is administered at a dose of 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, 1600 mg every 4 weeks, 2000 mg every 4 weeks, 2400 mg every 4 weeks, 2800 mg every 4 weeks, 3200 mg every 4 weeks, 3600 mg every 4 weeks, 4000 mg every 4 weeks, 4400 mg every 4 weeks, 4800 mg every 4 weeks, 5200 mg every 4 weeks, or 5600 mg every 4 weeks.
  • a brain MRI scan may be administered to the human subject to monitor/evaluate a human subject (e.g ., for ARIA-E or ARIA-H).
  • a brain MRI scan can be administered to the human subject to diagnose/evaluate/monitor adverse event(s) caused by administration of the antibody of the present disclosure.
  • the human subject is administered an MRI scan in between the administration of doses.
  • the human subject is administered an MRI scan before an increase in the dose of solanezumab.
  • the human subject is administered an MRI scan before administering a higher mg dose.
  • the human subject is administered an MRI scan before administering a dose of solanezumab.
  • centiloids reduction associated with amyloid plaques See , e.g., Klunk el al., “The Centiloid Project: Standardizing Quantitative Amyloid Plaque Estimation by PET,” Alzheimer’s & Dementia 11.1: 1-15 (2015) and Navitsky et al., “Standardization of Amyloid Quantitation with Florbetapir Standardized Uptake Value Ratios to the Centiloid Scale,” Alzheimer's & Dementia 14.12: 1565-1571 (2018), which are hereby incorporated by reference in their entireties.
  • the administration of solanezumab causes a reduction in the soluble Ab that is available in the brain. This reduction may be measured at about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, or about 72 weeks or about 80 weeks.
  • the administration of solanezumab results in a 5% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 10% lowering of the soluble Ab concentration.
  • administration of solanezumab results in a 15% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 20% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 25% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 30% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 35% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 40% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 45% lowering of the soluble Ab concentration.
  • administration of solanezumab results in a 50% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a greater than 50% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 55% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 60% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 65% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 70% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 75% lowering of the soluble Ab concentration.
  • administration of solanezumab results in an 80% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in an 85% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 90% lowering of the soluble Ab concentration.
  • the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline. In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline over a duration of about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or 76 weeks or about 80 weeks.
  • solanezumab is administered to the patient about every 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, about 76 weeks, or about 80 weeks.
  • the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline over a duration of 80 weeks.
  • the slowing of decline in the cognitive- functional composite endpoints from baseline is provided from the MMRM model or the Bayesian Disease Progression Model (DPM).
  • the antibody of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline.
  • the first or the second dose of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline in the cognitive- functional composite endpoints from baseline.
  • the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale (iADRS) from baseline. In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or 76 weeks.
  • iADRS Integrated Alzheimer's Disease Rating Scale
  • the present disclosure results in about 20 percent, about 25 percent, about 30 percent, about 32 percent, about 35 percent, about 40 percent, or about 45 percent slowing of decline in the Integrated Alzheimer's Disease Rating Scale from baseline.
  • the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of 76 weeks. In a particular embodiment, the present disclosure results in about 32 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of 76 weeks.
  • the antibody of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline.
  • the first or the second dose of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale (iADRS) from baseline.
  • the cognitive functional composite endpoint, including iADRS, of the subject is measured at about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or about 80 weeks.
  • solanezumab is administered to the patient who is a carrier of at least one allele of APOE e4 and causes at least a 5% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of at least one allele of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of at least one allele of APOE s4 and causes at least a 15% slowing of cognitive decline versus placebo on the MMSE scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 20% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 25% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the MMSE scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the MMSE scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 60% slowing of cognitive decline versus placebo on the MMSE scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the MMSE scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the CDS- SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 10% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the CDS-SB scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the CDS-SB scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the CDS-SB scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 50% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 55% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the CDS-SB scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the CDS-SB scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADCS-ADL scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADCS- ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADCS- ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADCS-iADL scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
  • solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the iADRS scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 10% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 15% slowing of cognitive decline versus placebo on the iADRS scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the iADRS scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the iADRS scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 55% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 60% slowing of cognitive decline versus placebo on the iADRS scale.
  • solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the iADRS scale.
  • the antibody of the present disclosure can be administered in simultaneous, separate, or sequential combination with an effective amount of a symptomatic agent to treat Alzheimer’s disease.
  • Symptomatic agents can be selected from cholinesterase inhibitors (ChEIs) and/or a partial /V-methyl-D-aspartate (NMDA) antagonists.
  • ChEIs cholinesterase inhibitors
  • NMDA partial /V-methyl-D-aspartate
  • the agent is a ChEI.
  • the agent is a NMDA antagonist or a combination agent comprising a ChEI and NMDA antagonist.
  • the disease characterized by Ab deposit in the brain of the subject is selected from preclinical Alzheimer’s disease, clinical AD, prodromal AD, mild AD, moderate AD, severe AD, Down’s syndrome, clinical cerebral amyloid angiopathy, or pre clinical cerebral amyloid angiopathy.
  • the subject is an early symptomatic AD patient.
  • the subject has prodromal AD and mild dementia due to AD.
  • the human subject has preclinical AD.
  • the human subject has evidence of elevated brain amyloid.
  • the subject has a genetic mutation that causes autosomal- dominant Alzheimer’s disease or at a higher risk for developing AD by virtue of carrying one or two APOE e4 alleles.
  • the subject carries one or two APOE s4 alleles, /. e. , the patient is heterozygous or homozygous.
  • biomarkers of a disease characterized by Ab deposits in the brain of a human subject, including Alzheimer’s disease.
  • biomarkers include, e.g ., amyloid deposits, amyloid plaque, Ab in CSF, Ab in the plasma, brain tau deposition, tau in plasma, or tau in cerebrospinal fluid and their use in screening, diagnosis, treatment, or prevention.
  • Non-limiting potential uses of such biomarkers include: 1) identification of subjects destined to become affected or who are in the “preclinical” stages of a disease; 2) reduction in disease heterogeneity in clinical trials or epidemiologic studies; 3) reflection of the natural history of disease encompassing the phases of induction, latency, and detection; and 4) target subjects for a clinical trial or for treatment/prevention of a disease.
  • the biomarkers may be used to assess whether a subject can be treated using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers may be used to assess whether a disease (as described herein) can be prevented in the subject using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers can be used to assess whether a subject is responsive to treatment or prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • the biomarkers can be used to stratify or classify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of diseases (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • the biomarkers may be used to assess disease state of a subject and/or the duration for administration of the antibodies or doses thereof, as described herein, to the subject.
  • the subject has a genetic mutation that causes autosomal- dominant Alzheimer’s disease or at a higher risk for developing AD by virtue of carrying one or two APOE e4 alleles.
  • the subject carries one or two APOE s4 alleles. Such patients, when administered solanezumab have better clinical outcomes than non-carriers.
  • the subject has low to moderate tau burden or has been determined to have low to moderate tau burden.
  • the subject has low to moderate tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18 F-flortaucipir) is from ⁇ 1.10 standardized uptake value ratio (SUVr) to ⁇ 1.46 SUVr.
  • the subject has low to moderate tau burden or has been determined to have low to moderate tau burden and carries one or two APOE e4 alleles.
  • the subject has very low tau burden or has been determined to have very low tau burden.
  • the subj ect has very low tau burden if the tau burden as measured by PET brain imaging (using, e.g ., 18 F-flortaucipir) is less than 1.10 SUVr.
  • the subject has very low tau burden or has been determined to have very low tau burden and carries one or two APOE e4 alleles.
  • the subject has very low to moderate tau burden or has been determined to have very low tau to moderate tau burden.
  • the subject has very low to moderate tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18 F- flortaucipir) is ⁇ 1.46 SUVr.
  • the subject has very low to moderate tau burden or has been determined to have very low to moderate tau burden and carries one or two APOE e4 alleles.
  • the subject does not have a high tau burden or has been determined to not have a high tau burden.
  • the human subject has high tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18 F- flortaucipir) is greater than 1.46 SUVr.
  • a subject with high tau is not administered the antibodies of the present disclosure.
  • the subject has does not have high tau burden or has been determined to not have a high tau burden and carries one or two APOE e4 alleles.
  • solanezumab, the dosing regimen, or the methods described the present disclosure is efficacious in human subjects having very low to moderate tau. In some embodiments, solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having low to moderate tau. In some embodiments, solanezumab is most efficacious in human subjects having a tau level i) less than or equal to about 1.14 SUVr or ii) from about 1.14 SUVr to about 1.27 SUVr.
  • solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having very low to moderate tau and carrying one or two APOE e4 alleles. In some embodiments, solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having low to moderate tau and carrying one or two APOE e4 alleles. In some embodiments, solanezumab is most efficacious in human subjects.
  • the tau level of a human subject can be determined by techniques and methods familiar to the diagnosing physician or a person of ordinary skill in the art.
  • a human subject who is suffering from a disease characterized by amyloid beta (Ab) deposits, is determined to have very low to moderate tau, low to moderate tau, or no high tau using techniques and methods familiar to the diagnosing physician or a person of ordinary skill in the art.
  • such methods can also be used to prescreen, screen, diagnose, evaluate increase or reduction in brain tau burden, and/or to assess the progress achieved in the treatment or prevention of the diseases described herein.
  • the methods can also be used to stratify subjects into groups and/or to identify which group of subjects is responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • the methods or techniques used to determine/detect tau level of a human subject can be used for prescreening or screening subjects and determining which subjects are responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • the tau level of a human subject can be determined using techniques or methods that, e.g ., detect or quantitate i) brain tau deposition, ii) tau in plasma, or iii) tau in cerebrospinal fluid.
  • brain tau burden, tau in plasma, or tau in cerebrospinal fluid can be used to stratify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of diseases (described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • Tau levels in the brain of human subject can be determined using methods, such as, tau imaging with radiolabeled PET compounds (Leuzy et al ., “Diagnostic Performance of R0948 F18 Tau Positron Emission Tomography in the Differentiation of Alzheimer Disease from Other Neurodegenerative Disorders,” JAMA Neurology 77.8:955-965 (2020); Ossenkoppele et al., “Discriminative Accuracy of 18 F-flortaucipir Positron Emission Tomography for Alzheimer Disease vs Other Neurodegenerative Disorders,” JAAJA 320, 1151-1162, doi:10.1001/jama.2018.12917 (2016), which are hereby incorporated by reference in their entireties.
  • the biomarker 18 F-flortaucipir which is a PET ligand
  • PET tau images can be, for example, quantitatively evaluated to estimate an SUVr (standardized uptake value ratio) by published methods (Pontecorvo et al ., “A Multicentre Longitudinal Study of Flortaucipir (18F) in Normal Ageing, Mild Cognitive Impairment and Alzheimer's Disease Dementia,” Brain 142: 1723-35 (2019); Devous et al.
  • flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal Intensity J. Nucl. Med. 59:944-951 (2016), which is hereby incorporated by reference in its entirety.
  • counts within a specific target region of interest in the brain e.g, multiblock barycentric discriminant analysis or MUBADA, see Devous et al., “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med.
  • a reference region wherein the reference region is, e.g, whole cerebellum, (wholeCere), cerebellar GM (cereCrus), atlas-based white matter (atlasWM), subject- specific WM (ssWM, e.g, using parametric estimate of reference signal intensity (PERSI), see Southekal et al., “Flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2016), which is hereby incorporated by reference in its entirety).
  • the reference region is, e.g, whole cerebellum, (wholeCere), cerebellar GM (cereCrus), atlas-based white matter (atlasWM), subject- specific WM (ssWM, e.g, using parametric estimate of reference signal intensity (PERSI), see Southekal et al., “Flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal
  • a preferred method of determining tau burden is a quantitative analysis reported as a standardized uptake value ratio (SUVr), which represents counts within a specific target region of interest in the brain (e.g, MUBADA,) when compared with a reference region (e.g, using PERSI).
  • phosphorylated tau P-tau; either phosphorylated at threonine 181 or 217) can be used to measure the tau load/burden for the purposes of the present disclosure (Barthelemy et al, “Cerebrospinal Fluid Phospho-tau T217 Outperforms T181 as a Biomarker for the Differential Diagnosis of Alzheimer's Disease and PET Amyloid positive Patient Identification,” Alzheimer’s Res. Ther.
  • the present disclosure includes, in some embodiments, the use of anti-tau antibodies disclosed in WO 2020/242963 to measure the tau load/burden in a subject.
  • the anti-tau antibodies disclosed in WO 2020/242963 are directed against isoforms of human tau expressed in the CNS (e.g ., recognizing the isoforms expressed in the CNS and not recognizing isoforms of human tau expressed exclusively outside the CNS).
  • Such antibodies against isoforms of human tau expressed in the CNS can be used in a method of identifying/selecting a patient as one or more of: (i) having a disease disclosed herein; (ii) at risk for having a disease disclosed herein; (iii) in need of treatment for a disease disclosed herein; or (iv) in need of neurological imaging.
  • a subject is positive for amyloid deposits when amyloid is detected in the brain by methods such as, amyloid imaging with radiolabeled PET compounds or using a diagnostic that detects Ab or a biomarker for Ab.
  • exemplary methods that can be used in the present disclosure to measure the brain amyloid load/burden include, e.g., Florbetapir (Carpenter, et al., “The Use of the Exploratory IND in the Evaluation and Development of 18 F-PET Radiopharmaceuticals for Amyloid Imaging in the Brain: A Review of One Company's Experience,” The Quarterly Journal of Nuclear Medicine and Molecular Imaging 53.4:387 (2009), which is hereby incorporated by reference in its entirety); Florbetaben (Syed et al, “[ 18 F]Florbetaben: A Review in b-Amyloid PET Imaging in Cognitive Impairment,” CNS Drugs 29, 605-613 (2015), which is hereby incorporated by reference in its entirety); and Flutemetamol (Heurling et al, “Imagin
  • 18 F-florbetapir can provide a qualitative and quantitative measurement of brain plaque load in patients, including patients with prodromal AD or mild AD dementia. For example, the absence of significant 18 ]-florbetapir signal on a visual read indicates patients clinically manifesting cognitive impairment have sparse to no amyloid plaques. As such, 18 F- florbetapir also provides a confirmation of amyloid pathology. 18 F-Florbetapir PET also provides quantitative assessment of fibrillar amyloid plaque in the brain and, in some embodiments, can be used to assess amyloid plaque reductions from the brain by antibodies of the present disclosure.
  • Amyloid imaging with radiolabeled PET compounds can also be used to determine if Ab deposit in the brain of a human patient is reduced or increased (e.g, to calculate the percentage reduction in Ab deposit post treatment or to assess the progression of AD).
  • a person of skill in the art can correlate the standardized uptake value ratio (SUVr) values obtained from amyloid imaging (with radiolabeled PET compounds) to calculate the % reduction in Ab deposit in the brain of the patient before and after treatment.
  • SUVr standardized uptake value ratio
  • the SUVr values can be converted to standardized centiloid (CL) units, where 100 is average for AD and 0 is average for young controls, allowing comparability amongst amyloid PET tracers, and calculation of reduction according to centiloid units (Klunk et al ., “The Centiloid Project: Standardizing Quantitative Amyloid Plaque Estimation by PET,” Alzheimer ’s & Dementia 11.1: 1-15 (2015) and Navitsky etal ., “Standardization of Amyloid Quantitation with Florbetapir Standardized Uptake Value Ratios to the Centiloid Scale,” Alzheimer's & Dementia 14.12: 1565-1571 (2016), which are hereby incorporated by reference in their entireties).
  • the change in brain amyloid plaque deposition from baseline is measured by 18 F-florbetapir PET scan.
  • Cerebrospinal fluid or plasma-based analysis of b-amyloid can also be used to measure the amyloid load/burden for the purposes of the present disclosure.
  • Ab42 can be used to measure brain amyloid (Palmqvist, S. et al ., “Accuracy of Brain Amyloid Detection in Clinical Practice Using Cerebrospinal Fluid Beta-amyloid 42: a Cross- validation Study against Amyloid Positron Emission Tomography. JAMA Neurol 71, 1282-1289 (2014), which is hereby incorporated by reference in its entirety).
  • the ratio of Ab42/Ab40 or Ab42/Ab38 can be used as a biomarker for amyloid beta (Janelidze etal ., “CSF Abeta42/Abeta40 and Abeta42/Abeta38 Ratios: Better Diagnostic Markers of Alzheimer Disease,” Ann Clin Transl Neurol 3, 154-165 (2016), which is hereby incorporated by reference in its entirety).
  • deposited brain amyloid plaque or Ab in CSF or plasma can be used to stratify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
  • solanezumab is administered by intravenous infusion. In another embodiment, solanezumab is administered subcutaneously.
  • Solanezumab binds selectively to Ab found in the brain.
  • Exemplary embodiments of anti-Ab antibodies of the present disclosure include solanezumab, which is described in (including methods of making and using it) in the following patent documents, which are expressly incorporated herein by reference: US Patent No. 7,195,761, US Patent Application Publication No. 20060039906, US Patent No. 7,892,545, US Patent No. 8,591,894, US Patent No, 7,771,722, US Patent Application Publication No. 20070190046.
  • solanezumab is an IgGl monoclonal antibody having complementarity-determining regions (CDRs). Solanezumab binds to the mid domain of the Ab peptide. In some embodiments, the antibody has the following sequence.
  • VVMTQ SPL SLP VTLGQP ASISCRS SQ SLIY SDGN AYLHWFLQKPGQ SPRLLIYK V SNRF S GVPDRF S GS GS GTDF TLKI SRVE AED VGV Y Y C S Q S THVP WTF GQGTK VEI KRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ E S VTEQD SKD STYSLSSTLTL SK AD YEKHK V Y ACE VTHQGL S SP VTK SFNRGEC
  • the antibody of the preceding sequence (as well the antibodies of the patents noted above in the following patent documents US Patent No. 7,195,761, US Patent Application Publication No. 20060039906, US Patent No. 7,892,545, US Patent No. 8,591,894, US Patent No, 7,771,722, US Patent Application Publication No. 20070190046) may be formulated and referred to as solanezumab.
  • the present embodiments include antibodies with SEQ ID NOs: 1 and 2 or other antibodies, as well as those antibodies which have been formulated in a composition called solanezumab.
  • any of the antibodies recited in the present disclosure may be used.
  • an “antibody” is an immunoglobulin molecule comprising two HC and two LC interconnected by disulfide bonds.
  • the amino terminal portion of each LC and HC includes a variable region responsible for antigen recognition via the complementarity determining regions (CDRs) contained therein.
  • CDRs complementarity determining regions
  • the CDRs are interspersed with regions that are more conserved, termed framework regions. Assignment of amino acids to CDR domains within the LCVR and HCVR regions of the antibodies of the present disclosure is based on the following: Rabat numbering convention (Rabat, et al ., Ann. NY Acad. Sci. 190:382-93 (1971); Rabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S.
  • the antibodies of the present disclosure are monoclonal antibodies (“mAbs”).
  • Monoclonal antibodies can be produced, for example, by hybridoma technologies, recombinant technologies, phage display technologies, synthetic technologies, e.g., CDR- grafting, or combinations of such or other technologies known in the art.
  • the monoclonal antibodies of the present disclosure are human or humanized. Humanized antibodies can be engineered to contain one or more human framework regions (or substantially human framework regions) surrounding CDRs derived from a non-human antibody.
  • Human framework germline sequences can be obtained from ImunoGeneTics (INGT) via their website (imgt.cines.fr), or from The Immunoglobulin FactsBook by Marie-Paule Lefranc and Gerard Lefranc, Academic 25 Press, 2001, ISBN 01244135 l.Techinques for generating human or humanized antibodies are well known in the art.
  • the antibody, or the nucleic acid encoding the same is provided in isolated form.
  • isolated refers to a protein, peptide or nucleic acid that is not found in nature and is free or substantially free from other macromolecular species found in a cellular environment.
  • substantially free means the protein, peptide or nucleic acid of interest comprises more than 80% (on a molar basis) of the macromolecular species present, preferably more than 90% and more preferably more than 95%.
  • Solanezumab may be administered as a pharmaceutical composition.
  • the pharmaceutical composition comprising an antibody of the present disclosure can be administered to a subject at risk for, or exhibiting, diseases or disorders as described herein by parental routes (e.g. , subcutaneous, intravenous, intraperitoneal, intramuscular). Subcutaneous and intravenous routes are preferred.
  • treatment include restraining, slowing, or stopping the progression or severity of an existing symptom, condition, disease, or disorder in a subject.
  • subject refers to a human subject or a patient.
  • prevention means prophylactic administration of the antibody of the present disclosure to an asymptomatic subject or a subject with pre-clinical Alzheimer’s disease to prevent onset or progression of the disease.
  • disease characterized by deposition of Ab or a “disease characterized by Ab deposits” are used interchangeably and refer to a disease that is pathologically characterized by Ab deposits in the brain or in brain vasculature. This includes diseases such as Alzheimer’s disease, Down’s syndrome, and cerebral amyloid angiopathy.
  • a clinical diagnosis, staging or progression of Alzheimer’s disease can be readily determined by the attending diagnostician or health care professional, as one skilled in the art, by using known techniques and by observing results.
  • the cognitive and functional assessment can be used to determine changes in a patient’s cognition (e.g, cognitive decline) and function (e.g, functional decline).
  • cognitive Alzheimer’s disease as used herein is a diagnosed stage of Alzheimer’s disease.
  • pre-clinical Alzheimer’s disease is a stage that precedes clinical Alzheimer’s disease, where measurable changes in biomarkers (such as CSF Ab42 levels or deposited brain plaque by amyloid PET) indicate the earliest signs of a patient with Alzheimer’s pathology, progressing to clinical Alzheimer’s disease. This is usually before symptoms such as memory loss and confusion are noticeable.
  • pre-clinical Alzheimer’s disease also includes pre-symptomatic autosomal dominant carriers, as well as patients with higher risk for developing AD by virtue of carrying one or two APOE s4 alleles.
  • a reduction or slowing of cognitive decline can be measured by cognitive assessments such as Clinical Dementia Rating - summary of boxes (CDR-SB), Mini-Mental State Exam (MMSE), or Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-Cog).
  • cognitive assessments such as Clinical Dementia Rating - summary of boxes (CDR-SB), Mini-Mental State Exam (MMSE), or Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-Cog).
  • a reduction or slowing of functional decline can be measured by functional assessments such as Alzheimer’s Disease Competence Scale- Activities of Daily Living (ADCS-ADL).
  • mg/kg means an amount, in milligrams, of antibody or drug administered to a subject based on his or her bodyweight in kilograms. A dose is given at one time. For example, a 10 mg/kg dose of antibody for a subject weighing 70 kg would be a single 700 mg dose of antibody given in a single administration. Similarly, a 20 mg/kg dose of antibody for a subject weighing 70 kg would be a 1400 mg dose of antibody given at a single administration.
  • a human subject has “very low tau” burden if the tau burden is less than 1.10 SUVr ( ⁇ 1.10 SUVr) using 18 F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (multiblock barycentric discriminant analysis or MUBADA, see Devous el al., “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18,” ./. Nucl. Med. 59:937-943 (2016)) when compared with a reference region (parametric estimate of reference signal intensity or PERSI, see , e.g.
  • a human subject has “very low tau to moderate tau” burden if the tau burden is less than or equal to 1.46 SUVr (i.e., ⁇ 1.46 SUVr) using 18 F -flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18,” J. Nucl. Med. 59:937-943 (2016)) when compared with a reference region (PERSI, see , Southekal et al.
  • a human subject has “low tau to moderate tau” burden if the tau burden is from greater than or equal to 1.10 to less than or equal to 1.46 (i.e., ⁇ 1.10 SUVr to ⁇ 1.46 SUVr) using 18 F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med. 59:937-943 (2016)) when compared with a reference region (PERSI, see , Southekal et al.
  • a human subject has “high tau” burden if the tau burden is greater than 1.46 SUVr (i.e., >1.46 SUVr) using 18 F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med.
  • compositions for treating the diseases or disorders described herein are equally applicable to use of a composition for treating the diseases or disorders described herein and/or compositions for use and/or uses in the manufacture of a medicaments for treating the diseases or disorders described herein.
  • Alzheimer’s patients were studied in two phase 3, double-blind trials (Expedition- 1 and Expedition-2, ClinicalTrials.gov numbers, NCT00905372 andNCT00904683, respectively), which randomly assigned 1012 and 1040 patients, respectively, with mild-to-moderate Alzheimer's disease to receive placebo or solanezumab (administered intravenously at a dose of 400 mg) every 4 weeks for 18 months.
  • Doody, etal. “Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer’s Disease”, New England Journal of Medicine, 370; 4, pp. 311-321 (2014), which is hereby incorporated by reference in its entirety).
  • the primary outcomes were the changes from baseline to week 80 in scores on the 11 -item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cogl l; range, 0 to 70, with higher scores indicating greater cognitive impairment) and the Alzheimer's Disease Cooperative Study- Activities of Daily Living scale (ADCS-ADL; range, 0 to 78, with lower scores indicating worse functioning).
  • ADCS-ADL Alzheimer's Disease Cooperative Study- Activities of Daily Living scale
  • ADAS-Cogl4 range, 0 to 90, with higher scores indicating greater impairment
  • Mild-to-moderate Alzheimer's disease was documented on the basis of a score of 16 to 26 on the Mini-Mental State Examination (MMSE; score range, 0 to 30, with higher scores indicating better cognitive function) and the criteria of the National Institute of Neurological and Communicative Disorders and Stroke- Alzheimer's Disease and Related Disorders Association.
  • MMSE Mini-Mental State Examination
  • the absence of depression was documented on the basis of a score of 6 or less on the Geriatric Depression Scale (score range, 0 to 15, with higher scores indicating more severe depression).
  • the primary outcome was the change from baseline to week 80 in the score on the 14-item cognitive subscale of the Alzheimer’s Disease Assessment Scale (ADAS-Cogl4; scores range from 0 to 90, with higher scores indicating greater cognitive impairment).
  • ADAS-Cogl4 Alzheimer’s Disease Assessment Scale
  • scores range from 0 to 90, with higher scores indicating greater cognitive impairment.
  • This trial included male and female patients, 55 to 90 years of age, who met the diagnostic criteria for probable Alzheimer’s disease according to the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association.
  • the Expedition-3 trial included only patients with mild Alzheimer’s disease who had biomarker evidence of amyloid-related disease, determined by means of either florbetapir positron-emission tomography (PET) scan or Ab1-42 measurements in cerebrospinal fluid (CSF).
  • PET florbetapir positron-emission tomography
  • CSF cerebrospinal fluid
  • Table 2 shows that administration of solanezumab to APOE e4 carriers of Expedition- 3 performed better in slowing decline than non-carriers:
  • Table 2 Change in MMSE based upon APOE e4 status.
  • Table 3 Change in Expedition-3 of CDS-SB by APOE e4 status.
  • the APOE s4 carriers had a -19% slowing of cognitive decline vs. placebo on the MMSE scale ( Figures 9 and 10A-B).
  • the carriers had a 18% slowing of cognitive decline vs. placebo on the CDR-SB scale ( Figures 11 and 12A-B).
  • the carriers had a 17% slowing of cognitive decline vs. placebo on the ADCS-ADL scale ( Figures 13 and 14A-B).
  • carriers had a 17% slowing of cognitive decline vs. placebo in Expedition-3 and a 28% slowing in pooled data from Expedition- 1 and -2 on the ADCS-iADL scale.
  • the APOE e4 carriers had a 15% slowing of cognitive decline vs. placebo in
  • Figure 15 shows a change on the iADRS scale between the carriers and non-carriers of APOE s4. Specifically, the carriers had a 11% slowing of cognitive decline vs. placebo in Expedition-3.
  • solanezumab slowed the clinical decline by 42% as measured with the ADAS- Cogl4 (p ⁇ 0.001) and by 28% in the ADCS-iADL (p ⁇ 0.01) in APOE e4 carriers.
  • solanezumab did not affect the clinical state of APOE e4 non-carriers.
  • Outcome measures show higher effect sizes for carriers compared to non-carriers and often larger and significant treatment differences in the carrier population.
  • carriers are younger and have higher amyloid loads and higher tau pathology.
  • the clinical decline across all scales for the placebo groups does not differ by carrier status.
  • LRP1 appears to facilitate tau internalization and degradation via an APOE e4 mediated mechanism.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present disclosure is directed to treatment or prevention of a disease characterized by deposition of Aβ in the brain using anti-Aβ antibodies. The diseases that can be treated or prevented include, e.g., Alzheimer's disease, Down's syndrome, and cerebral amyloid angiopathy. The invention, in some aspects, is related to doses and dosing regimens useful for such treatments. The invention is also related to, in some aspects, human subjects who are responsive to treatment or prevention of a disease characterized by Aβ in the brain using anti-Aβ antibodies. The invention is also related to patients who have one or more alleles of APOE e4.

Description

ANTI-AMYLOID BETA ANTIBODIES AND USES THEREOF
The present disclosure is related to methods of preventing or treating a disease with anti-amyloid beta antibodies, wherein the disease is characterized by deposition of amyloid beta (Ab) in a human subject. The present disclosure is also related to doses and dosing regimens of the anti-Ab antibodies useful for treating or preventing a disease characterized by deposition of Ab. Some aspects of the present disclosure are related to treating or preventing a disease characterized by deposition of Ab in human subjects, wherein the human subjects are selected, and/or treated with the antibodies of the present disclosure, based on the presence of one or two alleles of APOE e4 in the subject’s genome. The diseases that can be treated or prevented using antibodies, dosing regimens, or methods disclosed herein include, e.g ., Alzheimer’s disease (AD), Down’s syndrome, and cerebral amyloid angiopathy (CAA). In some embodiments, the present disclosure is related to slowing cognitive decline, functional decline, and/or memory loss in the subject. In some embodiments, the present disclosure is related to reducing amyloid load in the brain of the human subject.
BACKGROUND
A cure for AD is one of the most significant unmet needs of society. Accumulation of amyloid-b (Ab) peptide in the form of brain amyloid deposits/plaques is an early and essential event in Alzheimer’ s disease (AD) leading to neurodegeneration and consequently the onset of clinical symptoms, such as, cognitive and functional impairment (Selkoe, “The Origins of Alzheimer Disease: A is for Amyloid,” JAMA 283:1615-7 (2000); Hardy etal. , “The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics,” Science 297:353-6 (2002); Masters etal. , “Alzheimer’s Disease,” Nat. Rev. Dis. Primers 1:15056 (2015); and Selkoe et al, “The Amyloid Hypothesis of Alzheimer’s Disease at 25 years,” EMBO Mol. Med. 8:595-608 (2016)).
Amyloid beta (Ab) is formed by the proteolytic cleavage of a larger glycoprotein called amyloid precursor protein (APP). APP is an integral membrane protein expressed in many tissues, but especially in neuron synapses. APP is cleaved by g-secretase to release the Ab peptide, which encompasses a group of peptides ranging in size from 37-49 amino acid residues. Ab monomers aggregate into several types of higher order structures including oligomers, protofibrils, and amyloid fibrils. Amyloid oligomers are soluble and may spread throughout the brain, while amyloid fibrils are larger, insoluble, and can further aggregate to form amyloid deposits or plaques. The amyloid deposits found in human patients include a heterogeneous mixture of Ab peptides, some of which include N-terminal truncations and further may include N-terminal modifications such as an N-terminal pyroglutamate residue (pGlu).
The role for amyloid deposits in driving disease progression is supported by study of uncommon genetic variants that either increase or decrease Ab deposition (Fleisher et al ., “Associations Between Biomarkers and Age in the Presenilin 1 E280A Autosomal Dominant Alzheimer Disease Kindred: A Cross-sectional Study,” JAMA Neurol 72:316-24 (2015); Jonsson etal. , “A Mutation in APP Protects Against Alzheimer’s Disease and Age- related Cognitive Decline,” Nature 488:96-9 (2012)). In addition, the presence of amyloid deposits early in the disease increases the likelihood of progression of mild cognitive impairment (MCI) to AD dementia (Doraiswamy et al ., “Amyloid-b Assessed by Florbetapir F18 PET and 18-month Cognitive Decline: A Multicenter Study,” Neurology 79: 1636-44 (2012)). Interventions or therapies aiming at removal of Ab deposits (including amyloid plaques) are hypothesized to slow the clinical progression of AD.
Antibodies targeting amyloid are known, including, e.g., bapineuzumab, gantenerumab, aducanumab, GSK933776, solanezumab, crenezumab, ponezumab, and lecanemab (BAN2401). Some antibodies targeting amyloid have shown promise as a therapeutic for Alzheimer’s disease in preclinical studies. Despite this promise, most antibodies targeting amyloid have failed to meet therapeutic endpoints in multiple clinical trials. For example, solanezumab, an anti-Ab, has been studied in multiple Phase 3 clinical trials but did not meet its clinical endpoint. See Honig, et al. , “Trial of Solanezumab for Mild Dementia Due to Alzheimer’s Disease,” New England Journal of Medicine, vol 78, No. 4, pp. 321-300 (2018) (which is hereby incorporated by reference in its entirety). This article cites other Phase 3 trials of solanezumab that also failed to meet their clinical endpoints. Additional information about trials of solanezumab are published as Doody, et al. , “Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer’s Disease”, New England Journal of Medicine, 370; 4, pp. 311-321 (2014) (which is hereby incorporated by reference in its entirety). The history of anti-amyloid clinical trials spans almost two decades and has, for the most part, cast doubt on the potential of such therapies to effectively treat AD (Aisen, et al, “The Future of Anti-amyloid Trials,” The Journal of Prevention of Alzheimer's Disease 7:146-151 (2020); Budd, et al ., The Journal of Prevention of Alzheimer's Disease 4.4:255 (2017) and Klein, et al. , Alzheimer's Research & Therapy 11.1:101 (2019)).
Additionally, significant problems exist with long term chronic administration of Ab antibodies. Administration of Ab antibodies has led to adverse events in humans, such as, amyloid-related imaging abnormalities (ARIA), suggestive of vasogenic edema and sulcal effusions (ARIA-E), microhemorrhages and hemosiderin deposits (ARIA-H), infusion site reactions, and risk of immunogenicity. See , e.g. , Piazza and Winblad, “Amyloid-Related Imaging Abnormalities (ARIA) in Immunotherapy Trials for Alzheimer’s Disease: Need for Prognostic Biomarkers?” Journal of Alzheimer’s Disease, 52:417-420 (2016); Sperling, et al., “Amyloid-related Imaging Abnormalities in Patients with Alzheimer’s Disease Treated with Bapineuzumab: A Retrospective Analysis,” The Lancet Neurology 11.3: 241- 249 (2012); Brashear et al., “Clinical Evaluation of Amyloid-related Imaging Abnormalities in Bapineuzumab Phase III Studies,” J. of Alzheimer’s Disease 66.4:1409- 1424 (2018); Budd et al., “Clinical Development of Aducanumab, an Anti-Ab Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer's Disease,” The Journal of Prevention of Alzheimer's Disease 4.4: 255 (2017).
Although the exact cause of such adverse events is not known, it is believed that antibody treatment disrupts blood-brain barrier through interaction with the cerebral vascular amyloid and that this disruption leads to leaky barrier and the manifestation of edema in patients. Several mechanisms of action have been postulated, e.g, that removal of amyloid from the vessel wall destabilizes the neurovascular unit, localized inflammation/infiltrates in the neurovascular unit, increased levels of cerebral vascular amyloid due to higher levels of interstitial soluble Ab in response to parenchymal plaque clearance or altered localization of AQP-4 in astrocytic end feet projections in the neurovascular unit. Additionally, several therapeutic amyloid targeting antibodies have demonstrated dose-response related increases in ARIA-E. See, e.g, Brashear et al., “Clinical Evaluation of Amyloid-related Imaging Abnormalities in Bapineuzumab Phase III Studies,” J. of Alzheimer’s Disease 66.4:1409-1424 (2018); Budd et al., “Clinical Development of Aducanumab, an Anti-Ab Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer's Disease,” The Journal of Prevention of Alzheimer's Disease 4.4: 255 (2017). In some instances, there is a higher incidence rate of ARIA-E in patients harboring the epsilon-4 allele of apolipoprotein E (referred to herein as APOE s4).
To decrease the rate of ARIA-E adverse event while maintaining plaque clearance, some antibody treatment programs implement dose-titration schemes that include multiple dose escalations (3-4 steps) over a period of about 6-months prior to reaching their efficacious dose level. See , e.g, Budd et al ., “Clinical Development of Aducanumab, an Anti-Ab Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer's Disease,” The Journal of Prevention of Alzheimer's Disease 4.4:255 (2017) and Klein et al., “Gantenerumab Reduces Amyloid-b Plaques in Patients with Prodromal to Moderate Alzheimer’s Disease: a PET Substudy Interim Analysis,” Alzheimer's Research & Therapy 11.1:101 (2019). Such treatment regimens may not fully clear amyloid plaques or may delay clearance of amyloid plaque.
Furthermore, one of the challenges in treating Alzheimer’s disease is that it is still principally diagnosed and treated based on symptoms, e.g. , like a psychiatric illness, rather than based on brain pathology. This causes clinical trials to include heterogenous populations (e.g, with wide variation in levels of underlying pathology and/or different underlying diseases) which makes replicating clinical trial data very challenging. As such, determining whether subjects having Ab deposits may respond to an anti-Ab antibody treatment is uniquely challenging and the task of properly identifying whether a patient may respond to anti-Ab antibody treatments is of utmost importance for, e.g, a timely referral to a memory clinic, a correct and early AD diagnosis, initiation of symptomatic treatment, future planning, and initiating disease-modifying treatments.
Thus, a need exists for improved doses, dosing regimens, or methods that effectively treat/prevent AD and other forms of dementia, CAA and/or Down’s syndrome patients with anti-amyloid beta antibodies without causing or increasing problematic adverse events such as ARIA and ARIA-E. Additionally, a need exists for improved methods that properly identify whether a subject is going to respond to amyloid targeting therapeutics.
BRIEF DESCRIPTION OF DRAWINGS
“Fig.” denotes Figure.
Fig. 1 shows a table describing the effect size for APOE e4 carrier vs. non-carrier across Expedition-3 and Trailblazer-1 Clinical Trials. Abbreviations: Statistically sig = Statistically significant (p<0.05) Worse = performed worse than placebo; iADRS = Integrated Alzheimer's Disease (AD) Rating Scale; ADAS-Cog = Alzheimer's Disease Assessment Scale cognitive subscale; iADL = instrumental Activities of Daily Living; CDR-SB = Clinical Dementia Rating scale - sum of boxes; MMSE = Mini-Mental State Exam; ADAS = Alzheimer's Disease Assessment Scale; FAQ = Function Activities Questions.
Fig. 2 shows the effect size for APOE e4 carrier vs. non-carrier. Effect sizes of Mild AD in Sola (not amyloid selected). Abbreviations: Statistically sig = Statistically significant; Worse = performed worse than placebo; NA = Not available; ADAS-Cog-14 = 14-item Alzheimer's Disease Assessment Scale - Cognitive subscale; MMSE = Mini- Mental State Exam; CDR-SB = Clinical Dementia Rating scale - sum of boxes; ADL = Activities of Daily Living.
Fig. 3 shows change in cognition ADAS-Cogl4 (primary endpoint) from the Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADAS- Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE= Standard Error.
Fig. 4A shows change in cognition ADAS-Cogl4 by APOE e4 status from the Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, patients that are carriers of APOE e4. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. The APOE e4 carrier status-by-treatment interaction showed a p-value = 0.157. Abbreviations: AD= Alzheimer’ s disease; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE=Standard Error; APOE = Apolipoprotein E.
Fig. 4B shows change in cognition ADAS-Cogl4 by APOE e4 status from Expedition- 3 Clinical Trial of Solanezumab initiated in patients with mild AD dementia, patients that are not carriers of APOE e4. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. The APOE e4 carrier status- by-treatment interaction showed a p-value = 0.157. Abbreviations: AD= Alzheimer’ s disease; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE=Standard Error; APOE = Apolipoprotein E. Fig. 5A shows change in cognition ADAS-Cogl4 by APOE e4 Status. Pooled Expedition-1 and Expedition-2 Clinical Trial patients with Solanezumab in Mild AD Dementia, APOE e4 carriers. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE=Standard Error; APOE = apolipoprotein E.
Fig. 5B shows change in cognition ADAS-Cogl4 by APOE e4 status. This data related to pooled Expedition- 1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 non-carriers. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE=Standard Error; APOE = apolipoprotein E.
Fig. 6 shows change in complex ADLs - ADCS-iADL in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADL= Activities of Daily Living; ADCS- iADL=AD Cooperative Study -Instrumental Activities of Daily Living; LS=least squares; n=number; SE=Standard Error.
Fig. 7A shows change in ADLs - ADCS-iADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers. The APOE e4 carrier status-by-treatment interaction displayed a p-value = 0.878. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error.
Fig. 7B shows change in ADLs - ADCS-iADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non carriers. The APOE e4 carrier status-by-treatment interaction displayed a p-value = 0.878. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADCS- iADL=AD Cooperative Study -Instrumental Activities of Daily Living; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error.
Fig. 8A shows change in ADCS-iADLs by APOE e4 status in pooled Expedition-1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADCS- iADL=AD Cooperative Study -Instrumental Activities of Daily Living; LS=least squares; SE=Standard Error; n=number.
Fig. 8B shows change in ADCS-iADLs by APOE e4 status in pooled Expedition- 1 and Expedition-2 clinical trial patients with solanezumab in mild AD dementia, APOE e4 non carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADCS- iADL=AD Cooperative Study -Instrumental Activities of Daily Living; LS=least squares; SE=Standard Error; n=number.
Fig. 9 shows change in cognition-MMSE in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; LS=least squares; MMSE=Mini-Mental State Examination; n=number; SE=Standard Error.
Fig. 10A shows change in cognition-MMSE by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers. The APOE s4 carrier status-by- treatment interaction displayed a p-value of 0.201. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; MMSE=Mini-Mental State Examination; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error.
Fig. 10B shows change in cognition-MMSE by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non-carriers. The APOE e4 carrier status-by-treatment interaction displayed a p-value of 0.201. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; MMSE=Mini-Mental State Examination; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error.
Fig. 11 shows change in composite scale CDR-SB in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; CDR-SB=Clinical Dementia Rating Sum of Boxes; LS=least squares; n=number; SE=Standard Error.
Fig. 12A shows change in composite scale CDR-SB by APOE e4 status in Expedition- 3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; CDR- SB=Clinical Dementia Rating Sum of Boxes; LS=least squares; n=number; SE=Standard Error; APOE/Apoe = apolipoprotein.
Fig. 12B shows change in composite scale CDR-SB by APOE e4 status in Expedition- 3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non carriers. Patients could continue stable standard of care for AD, including drug and non drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; CDR- SB=Clinical Dementia Rating Sum of Boxes; LS=least squares; n=number; SE=Standard Error; APOE/Apoe = apolipoprotein.
Fig. 13 shows change in ADCS-ADL in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADL= Activities of Daily Living; ADCS-ADL=AD Cooperative Study-Activities of Daily Living; LS=least squares; n=number; SE=Standard Error.
Fig. 14A shows change in ADCS-ADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 carriers. The APOE s4 carrier status-by-treatment interaction displayed a p-value of 0.714. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error. Fig. 14B shows change in ADCS-ADL by APOE e4 status in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia, APOE e4 non-carriers. The APOE s4 carrier status-by-treatment interaction displayed a p-value of 0.714. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; APOE=apolipoprotein E; SE=Standard Error.
Fig. 15 shows change in composite scale: iADRS in Expedition-3 clinical trial of solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; iADRS=Integrated AD Rating Scale; LS=least squares; n=number; SE=Standard Error.
Fig. 16 shows change in cognition and ADLs at 80 weeks in Expedition-3 clinical trial of Solanezumab initiated in patients with mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADL= Activities of Daily Living; ADAS- Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; ADCS-ADL=AD Cooperative Study-Activities of Daily Living; CDR-SB=Clinical Dementia Rating Sum of Boxes; FAQ= Function Activities Questionnaire; iADRS=Integrated AD Rating Scale; LS=least squares; n=number of patients with evaluable 80-Week scale data; N/A=not applicable; MMSE=Mini-Mental State Examination; SE=standard error.
Fig. 17 shows baseline demographics pooled from Expedition- 1 and Expedition-2 clinical trials of Solanezumab in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: * = Based on number of patients with available APOE status (placebo N=614; solanezumab N=595); AD= Alzheimer’s disease; n=number; SD=standard deviation; APOE=Apolipoprotein E.
Fig. 18 shows baseline clinical characteristics of Expedition-3 clinical trial of solanezumab initiated in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AChEI=acetylcholinesterase inhibitor; ADCS-ADL=AD Cooperative Study-Activities of Daily Living; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; CDR- SB=Clinical Dementia Rating Sum of Boxes; FAQ= Function Activities Questionnaire; MMSE=Mini-Mental State Examination; n=number; NA=not available from topline results.
Fig. 19 shows baseline clinical characteristics pooled from Expedition- 1 and Expedition-2 clinical trials of solanezumab in mild AD dementia. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD=Alzheimer’ s disease; AChEI=acetyl cholinesterase inhibitor; ADCS-ADL=AD Cooperative Study-Activities of Daily Living; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; CDR-SB=Clinical Dementia Rating Sum of Boxes; FAQ= Function Activities Questionnaire; n=number; MMSE=Mini-Mental State Examination; N/A=not applicable; SD=standard deviation.
Fig. 20A shows change in complex ADLs: ADCS-iADL from Expedition-1 clinical Trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease;
ADL=Activities of Daily Living; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; SE=Standard Error.
Fig. 20B shows change in complex ADLs: ADCS-iADL from Expedition-2 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease;
ADL=Activities of Daily Living; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; SE=Standard Error.
Fig. 20C shows change in complex ADLs: ADCS-iADL pooled from Expedition-1 and Expedition-2 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADL= Activities of Daily Living; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; SE= Standard Error.
Fig. 20D shows change in complex ADLs: ADCS-iADL from Expedition-3 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease;
ADL=Activities of Daily Living; ADCS-iADL=AD Cooperative Study-Instrumental Activities of Daily Living; LS=least squares; n=number; SE=Standard Error. Fig. 21A shows change in cognition: ADAS-Cogl4 from Expedition-1 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADAS- Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE= Standard Error.
Fig. 21B shows change in cognition: ADAS-Cogl4 from Expedition-2 clinical trial. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADAS- Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE= Standard Error.
Fig. 21C shows change in cognition: ADAS-Cogl4 pooled from Expedition-1 and -2 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’s disease; ADAS-Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE=Standard Error.
Fig. 21D shows change in cognition: ADAS-Cogl4 pooled from Expedition-3 clinical trials. Patients could continue stable standard of care for AD, including drug and non-drug treatments, throughout the study. Abbreviations: AD= Alzheimer’ s disease; ADAS- Cogl4=AD Assessment Scale-Cognitive 14-item Subscale; LS=least squares; n=number; SE= Standard Error.
DETAILED DESCRIPTION
One aspect of the present disclosure provides for doses and dosing regimens of solanezumab circumventing problematic adverse events, such as ARIA with vasogenic edema, which have been observed in patients receiving therapeutic antibodies that bind to deposited amyloid and has been dose limiting for clinical development programs. Additionally, aspects of the present disclosure provide improved methods for identifying patients for treatment of AD and other forms of dementia, CAA and/or Down’s syndrome patients with anti-amyloid beta antibodies.
Doody et al., New England Journal of Medicine, 370; 4, pp. 311-321 (2014) (which is hereby incorporated by reference in its entirety) indicates that “[n]o clear differential treatment effects on efficacy measures were observed between APOE e4 carriers and noncarriers” in the Expedition-1 study. However surprisingly, it has now been found that administering solanezumab to a human subject that has one or two alleles of APOE e4 (e.g, a carrier of APOE e4) provides unexpected and surprising efficacy when compared to non carriers of one or more of those alleles. Thus, the present disclosure involves administering doses of solanezumab or other anti-Ab antibodies to patients who have one or two alleles of APOE s4 as a means of treating patients with AD, preventing AD, and/or slowing cognitive/functional decline in patients.
Specifically, it has been found that there is a greater effect in carriers of APOE e4 than in non-carriers when the patients are administered anti-Ab antibodies. This means that the patients that have APOE e4 have less cognitive decline than non-carriers, when measured using various clinical measurements and at various endpoints. In fact, as shown herein, the data from Expedition- 1 and Expedition-2 trials, when combined, show that there is a surprising difference in cognitive outcomes for carriers vs. non-carriers of APOE e4.
Another aspect of the present disclosure is based on the discovery that Alzheimer’s patients having one or two alleles of APOE e4 are more responsive to treatment with anti- N3pGlu Ab antibodies as compared to non-carriers of APOE e4. Yet another aspect of the present disclosure is based on the discovery that Alzheimer’s patients having one or two alleles of APOE e4 and low or moderate tau, very low to moderate tau, or not having high tau are responsive to treatment with anti-N3pGlu Ab antibodies.
In some embodiments, the patients can be stratified/identified/selected/treated based on the amount of tau present in the subject’s brain (e.g, in the whole brain or in portions of the brain) and the presence of one or two alleles of APOE e4. In some embodiments, the patients can be stratified/identified/selected/treated based on the stages of AD progression (e.g. , based on the spread of tau in the brain) and the presence of one or two alleles of APOE s4.
Some aspects of the present disclosure provide for a dosing regimen where a human subject, suffering from a disease characterized by Ab deposits in their brain, is administered solanezumab in one step or two steps. In some embodiments, the human subject is administered one or more doses of the antibodies of the present disclosure, optionally, once about every 4 weeks. In some embodiments, the human subject is administered one or more doses of solanezumab in a first step, wherein each dose is administered once about every 4 weeks. About four weeks after administering the one or more first doses, the human subject is administered one or more second doses in a second step.
Some aspects of the present disclosure are related to identifying the stage/progression of AD in a patient based on i) the global or overall tau burden in the brain of a human subject or ii) the spread of tau in the subject’s brain or portions thereof. See, e.g ., International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety. The stratification of patients based on amount of tau in the brain or AD progression in portions of brain can be used to determine, e.g. , whether a patient will respond to treatments. Stratification/selection of patient population based on amount of tau in the brain or AD progression in portions of brain is also helpful in solving the patient heterogeneity and replicability problems faced during design and performance of clinical trials. See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
In some embodiments, the patients can be stratified/identified/selected/treated based on the amount of tau present in the subject’s brain (e.g, in the whole brain or in portions of the brain). See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety. In other embodiments, the patients are stratified/identified/selected/treated based on stages of AD progression (e.g, based on the spread of tau in the brain). For example, during some stages, tau burden in an AD patient is isolated to frontal lobe or regions of the temporal lobe that do not include the posterolateral temporal region (PLT). Another stage of AD is where tau burden in an AD patient is limited to the posterolateral temporal (PLT) or occipital regions. Yet another stage of AD is when the tau burden in an AD patient is present in the parietal or precuneus region or in the frontal region along with tau burden in PLT or occipital regions. See, e.g, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
Other aspects of the present disclosure provide for human subjects that are responsive to treatment or prevention of a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject. In some embodiments, of this aspect of the present disclosure, the responsive human subjects include human subjects having low to moderate tau burden or very low to moderate tau burden, optionally having one or more alleles of APOE e4. In some embodiments, of this aspect of the present disclosure, the responsive human subjects exclude human subjects with high tau burden, optionally having one or more alleles of APOE s4. In some embodiments, solanezumab is administered to the responsive human subjects for treatment or prevention of a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject. See, e.g ., International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
Some aspects of the present disclosure are related to a method of treating or preventing a disease characterized by Ab deposits in the brain of a human subject wherein the human subject is clinically asymptomatic. This method includes administering to an APOE e4 carrier (e.g, one who has one or more of the APOE e4 alleles) a dose of solanezumab. In terms of solanezumab the dose may be 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, or 1600 mg every 4 weeks. Of course, other doses may be used. This may be giving an initial dose of 400 mg, and either maintaining the patient at 400 mg or titrating up to 800 mg every 4 weeks, or titrating up 1200 mg every 4 weeks, or titrating up 1600 mg over time. Other embodiments may involve giving an initial dose of 1600 mg and then maintaining at that dose or titrating down to 400 mg, 800 mg, or 1200 mg. Those skilled in the art will appreciate how to titrate up or down the solanezumab dose, or to maintain the patient on a particular dose (and the timing associated with making a dosing change).
Those skilled in the art will appreciate that changing the dose of solanezumab may be based upon numerous factors, including PET scans, clinical observations, performance by the patient on various “tests,” etc.
In some embodiments, the clinically asymptomatic subjects are known to have an Alzheimer's disease-causing genetic mutation. In the present disclosure, “clinically asymptomatic subjects known to have an Alzheimer's disease-causing genetic mutation” include patients known to have a PSEN1 E280A Alzheimer's disease-causing genetic mutation (Paisa mutation), a genetic mutation that causes autosomal-dominant Alzheimer's disease or are at higher risk for developing AD by virtue of carrying one or two APOE e4 alleles.
As used herein, “anti-Ab antibody" refers to an antibody that binds to an epitope present on Ab. In some embodiments, the anti-Ab antibody binds to a soluble form of Ab. In other embodiments, the anti-Ab antibody binds to an insoluble form of Ab, such as Ab plaques. In some embodiments, the anti-Ab antibody binds an epitope present in Ab1-40 or Abΐ- 42. In other embodiments, the anti-Ab antibody binds an epitope present in a truncated form of Ab1-40 or Ab1-42, for example, a truncated form lacking 1-20 N terminal amino acids and/or lacking 1-20 C-terminal amino acids. In other embodiments, the anti-Ab antibody binds an epitope present in a fragment of Ab1-40 or Ab1-42 and having a length of about 5-20 amino acids. Anti-Ab antibodies have been disclosed in the art. (See, e.g., U S. Patent Nos. 10,851,156; 10,738,109; 10,662,239; 10,654,917; 10,647,759; 10,603,367; 10,519,223; 10,494,425; 10,464,976; 10,112,991; 10,112,987; 10,035,847; 9,944,696; 9,939,452; 9,895,429; 9,834,598; 9,738,712; 9,585,956; 9,573,994; 9,382,312; 9,329,189; 9,309,309; 9,309,307; 9,272,031; 9,181,332; 9,176,150; 9, 175, 094;9, 146,244; 9,133,267; 9,125,846; 9,062,102; 9,051,364; 9,051,363; 8,916,165; 8,906,370; 8,906,367; 8,889,138; 8,796,439; 8,795,664; 8,710,193; 8,636,981; 8,614,299; 8,591,894; 8,507,206; 8,491,903; 8,470,321; 8,425,905; 8,420,093; 8,414,893; 8,398,978; 8,383,113; 8,337,848; 8,333,967; 8,323,654; 8,303,954; 8,268,973; 8,268,593; 8,246,954; 8,227,576; 8,222,002; 8,221,750; 8,173,127; 8,128,930; 8,128,928; 8,124,353; 8,124,076; 8,106,164; 8,105,594; 8,105,593; 8,025,878; 7,955,812; 7,939,075; 7,932,048; 7,927,594; 7,906,625; 7,902,328; 7,893,214; 7,892,545; 7,892,544; 7,871,615; 7,811,563; 7,807,165; 7,807,157; 7,790,856; 7,780,963; 7,772,375; 7,763,250; 7,763,249; 7,741,448; 7,731,962; 7,700,751; 7,625,560; 7,582,733; 7,575,880; 7,339,035; 7,320,790; 7,318,923; 7,256,273; 7,195,761; 7,189,819; 7,179,892; 7,122,374; 7,060,270; 6,815,175; 6,787,637; and 6,750,324; which are hereby incorporated by reference in their entireties).
One aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having an APOE e4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subj ect that has one or two alleles of APOE e4. In some embodiments, the antibody administered to the human subject is an anti-Ab antibody.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject. In some embodiments, the antibody administered to the human subject is an anti-Ab antibody.
Yet another aspect of the present disclosure is related to method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE s4 allele; and ii) if the human subject is identified as having an APOE e4 allele, administering or having administered a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject. In some embodiments, the antibody administered to the human subject is an anti-Ab antibody.
In one aspect the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE s4 allele. In some embodiments, the antibody administered to the human subject is an anti-Ab antibody.
One aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject having an APOE e4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subject that has one or two alleles of APOE e4.
Another aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
An aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE e4 allele; and ii) if the human subject is identified as having an APOE s4 allele, administering or having administered an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
Another aspect of the present disclosure is related to a method of reducing amyloid load in the brain of a human subject, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE s4 allele.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising administering to the human subject an effective amount of solanezumab, wherein the human subject has been determined as having a very low to moderate tau burden or low to moderate tau burden. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have one or two alleles of APOE e4 and very low to moderate tau burden or low to moderate tau burden comprising: administering to the human subject an effective amount of solanezumab. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
In one aspect, the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined as not having a high tau burden comprising: administering to the human subject an effective amount of solanezumab. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has low to moderate tau burden or a very low to moderate tau burden; and if the human subject has low to moderate tau burden or a very low to moderate tau burden, then: administering to the human subject an effective amount of solanezumab. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising administering to the human subject an effective amount of solanezumab, wherein the human subject has been determined as not having a high tau burden. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has high tau burden; and if the human subject does not have high tau burden, then: administering to the human subject an effective amount of solanezumab. In some embodiments of this aspect of the present disclosure, the human subject has one or two alleles of APOE e4.
In some aspects, an anti-Ab antibody may be used to decrease, prevent further increase of tau burden, or slow the rate of tau accumulation in different portions of a human brain, e.g ., in different lobes of the brain of a human subject. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the frontal lobe of the human brain. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the parietal lobe of the human brain. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the occipital lobe of the human brain. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the temporal lobe of the human brain. In some embodiments, solanezumab is used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in the posterolateral temporal lobe.
An aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the temporal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the temporal lobe of the brain and administering an anti-Ab to the human subject. In some embodiments, the human subject has tau burden in the posterolateral temporal lobe.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the occipital lobe of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the occipital lobe of the brain and administering an anti-Ab to the human subject.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject who has been determined to have tau burden in the parietal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the parietal lobe of the brain and administering an anti-Ab to the human subject.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in the frontal lobe of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the frontal lobe of the brain and administering an anti-Ab to the human subject. Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in the posterolateral temporal (PLT) and/or occipital lobe of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject comprising determining whether the human subject has tau burden in the posterolateral temporal (PLT) and/or occipital lobe of the brain and administering an anti-Ab to the human subject.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden in i) parietal or precuneus region or ii) in frontal region along with tau burden in PLT or occipital regions of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits comprising determining whether the human subject has tau burden in i) parietal or precuneus region or ii) in the frontal region along with tau burden in PLT or occipital regions of the brain and administering an anti-Ab to the human subject.
Another aspect of the present disclosure is related to a method of treating or preventing a disease characterized by amyloid beta deposits in the brain of a human subject who has been determined to have tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain wherein the method comprises administering an anti-Ab to the human subject. Another aspect of the invention is related to a method of treating or preventing a disease characterized by amyloid beta deposits comprising determining whether the human subject has tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain and administering an anti-Ab to the human subject.
In some aspects, the present disclosure is related to a method of selecting a human subject for treatment or prevention of a disease characterized by amyloid beta deposits in the brain of a human subject. In some embodiments, the human subject is selected based on the amount of global (overall) tau in the brain of the human subject. For example, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has very low to moderate tau in the brain. In another embodiment, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has low to moderate tau (or intermediate tau) in the brain. In another embodiment, the human subject is excluded from treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has high tau in the brain. In some embodiments, the human subject is selected based on progression of AD in the brain of the human subject. For example, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the frontal lobe of the brain. In another embodiment, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the parietal lobe of the brain. In another embodiment, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the occipital lobe of the brain. In another embodiment, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the temporal lobe of the brain. In some embodiments, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in the posterolateral temporal (PLT) and/or occipital lobe of the brain. In some embodiments, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden present in i) parietal or precuneus region or ii) in frontal region along with tau burden in PLT or occipital regions of the brain. In some embodiments, the human subject is selected for treatment or prevention of a disease characterized by amyloid beta deposits in the brain because the patient has tau burden i) isolated to frontal lobe or ii) in regions of the temporal lobe that do not include the posterolateral temporal region (PLT) of the brain.
In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have a posterior-lateral temporal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe and occipital lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, and parietal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe, and frontal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe and / or frontal lobe tau burden. In some embodiments, the subject described in the various aspects of the present disclosure has been determined to have posterior-lateral temporal lobe, occipital lobe, parietal lobe and / or frontal lobe tau burden corresponds a neurological tau burden of greater than 1.46 SUVr based on PET imaging. In some embodiments, tau burden in a portion of the human brain ( e.g ., in a lobe of the brain) can be used to determine whether administration of the anti-Ab should be discontinued. For instance, decrease, prevention of further increase, or slowing in the rate of tau burden/accumulation in a portion of the brain can be used as metric to determine the duration of administration of solanezumab. In some embodiments, solanezumab is administered to the subject until there is a decrease, prevention of further increase, or slowing in the rate of tau burden/accumulation in the temporal lobe, the occipital lobe, the parietal lobe, or the frontal lobe.
In some embodiments, the tau burden present in a portion of the brain of a human subject can be used for selection of optimal treatment regimens or for administration of therapeutic modalities in combination with solanezumab. For example, the presence of tau burden in the frontal lobe of the brain of an amyloid positive human subject can be used as a metric to determine whether the human subject will benefit from administration of an anti-Ab antibody alone or its combination with an anti-tau antibody. In some embodiments, solanezumab in combination with an anti-tau antibody may be used to decrease, prevent further increase, or slow the rate of tau burden/accumulation in different portions of a human brain, e.g., in different lobes of the brain of a human subject. In some embodiments, the tau burden in different portions of a human brain, e.g, in different lobes of the brain of a human subject can be used for i) tracking patient’s response to treatment, or ii) when a therapy may need to be reinitiated.
In some embodiments, the antibodies, methods, or dosing regimens described in various aspects of the present disclosure cause: i) reduction in Ab deposits in the brain of the human subject and/or ii) slow cognitive decline or functional decline in the human subject. In some embodiments, the antibodies, methods, or dosing regimens described herein results in reduction of amyloid plaques.
Table 1 below shows how administration of solanezumab affects the amount of tau in various lobes of the brain, and how that amount differs based upon whether the patient is an APOE e4 carrier or non-carrier.
Table 1:
Figure imgf000024_0001
Figure imgf000025_0001
In some embodiments, solanezumab is administered to the subject for a duration sufficient to treat or prevent the disease. In some embodiments, solanezumab (including the first doses of the antibody and the second doses of the antibody) is administered to the subject for a duration of up to about 54 weeks, about 72 weeks, or about 80 weeks, optionally, about once every 4 weeks or about once every month.
In some embodiments, the antibody of the present disclosure is administered to the patient till amyloid levels in the brain of the patient reach a normal range. In some embodiments, the antibody of the present disclosure is administered to the subject till amyloid deposits or plaques are cleared from the subj ecf s brain. In some embodiments, the antibody is administered to the subject till the subject reaches amyloid negative status (defined as <24.1 CL amyloid plaque). In the present disclosure, the term “normal range” of amyloid plaque in brain is used interchangeably with brain amyloid plaque is “cleared.” The normal range of amyloid plaque is defined as demonstrating an amyloid plaque level of 25 centiloids or lower for two consecutive PET scans at least 6 months apart or a single PET scan demonstrating a plaque level of less than 11 centiloids.
In some embodiments, the dose of solanezumab is every 4 weeks (possibly modifying the dose every 4 weeks, as outlined herein) for up to about 54 weeks, about 72 weeks, or about 80 weeks. In some embodiments, solanezumab slows disease progression in patients with early symptomatic Alzheimer’s disease (AD) and with the presence of intermediate brain tau burden. In some embodiments, the antibody of the present disclosure is administered to the subject as one dose or more than one doses. In some embodiments, the patients receive solanezumab at a dose of 400 mg given intravenously every 4 weeks for 76 weeks or 80 weeks or some other time period. In some embodiments, the dose of solanezumab may be increased to 800 mg, 1200 mg, or 1600 mg given every 4 weeks. In some embodiments, the antibody of the present disclosure is administered at a dose of 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, 1600 mg every 4 weeks, 2000 mg every 4 weeks, 2400 mg every 4 weeks, 2800 mg every 4 weeks, 3200 mg every 4 weeks, 3600 mg every 4 weeks, 4000 mg every 4 weeks, 4400 mg every 4 weeks, 4800 mg every 4 weeks, 5200 mg every 4 weeks, or 5600 mg every 4 weeks.
A brain MRI scan may be administered to the human subject to monitor/evaluate a human subject ( e.g ., for ARIA-E or ARIA-H). In some embodiments, a brain MRI scan can be administered to the human subject to diagnose/evaluate/monitor adverse event(s) caused by administration of the antibody of the present disclosure. In some embodiments, the human subject is administered an MRI scan in between the administration of doses. In some embodiments, the human subject is administered an MRI scan before an increase in the dose of solanezumab. In some embodiments, the human subject is administered an MRI scan before administering a higher mg dose. In some embodiments, the human subject is administered an MRI scan before administering a dose of solanezumab.
Those skilled in the art will appreciate how to measure centiloids reduction associated with amyloid plaques. See , e.g., Klunk el al., “The Centiloid Project: Standardizing Quantitative Amyloid Plaque Estimation by PET,” Alzheimer’s & Dementia 11.1: 1-15 (2015) and Navitsky et al., “Standardization of Amyloid Quantitation with Florbetapir Standardized Uptake Value Ratios to the Centiloid Scale,” Alzheimer's & Dementia 14.12: 1565-1571 (2018), which are hereby incorporated by reference in their entireties.
In some embodiments, the administration of solanezumab causes a reduction in the soluble Ab that is available in the brain. This reduction may be measured at about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, or about 72 weeks or about 80 weeks. In some embodiments, the administration of solanezumab results in a 5% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 10% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 15% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 20% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 25% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 30% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 35% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 40% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 45% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 50% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a greater than 50% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 55% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 60% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 65% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 70% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 75% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in an 80% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in an 85% lowering of the soluble Ab concentration. In other embodiments, administration of solanezumab results in a 90% lowering of the soluble Ab concentration.
Those skilled in the art will appreciate how to measure the concentration of the soluble Ab concentration. See , e.g., Siemers ER, Friedrich S, Dean RA, etal., “Safety and changes in plasma and cerebrospinal fluid amyloid b after a single administration of an amyloid b monoclonal antibody in subjects with Alzheimer disease,” Clin. Neuropharmacol. 2010; 33:67-73; and Farlow M, Arnold SE, van Dyck CH, et al ., “Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease,” Alzheimer ’s Dement 2012; 8:261- 71 (both of which are expressly incorporated herein by reference in their entirety).
In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline. In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline over a duration of about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or 76 weeks or about 80 weeks.
In some embodiments, solanezumab is administered to the patient about every 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, about 76 weeks, or about 80 weeks.
In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline over a duration of 80 weeks. In some embodiments, the slowing of decline in the cognitive- functional composite endpoints from baseline is provided from the MMRM model or the Bayesian Disease Progression Model (DPM). In some embodiments, the antibody of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline in the cognitive-functional composite endpoints from baseline. In some embodiments, the first or the second dose of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline in the cognitive- functional composite endpoints from baseline.
In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale (iADRS) from baseline. In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or 76 weeks.
In some embodiments, the present disclosure results in about 20 percent, about 25 percent, about 30 percent, about 32 percent, about 35 percent, about 40 percent, or about 45 percent slowing of decline in the Integrated Alzheimer's Disease Rating Scale from baseline.
In some embodiments, the present disclosure results in about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of 76 weeks. In a particular embodiment, the present disclosure results in about 32 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline over a duration of 76 weeks. In some embodiments, the antibody of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale from baseline. In some embodiments, the first or the second dose of the present disclosure is administered to the subject till it reaches about 15 to about 45 percent slowing of decline on the Integrated Alzheimer's Disease Rating Scale (iADRS) from baseline.
In some embodiments, the cognitive functional composite endpoint, including iADRS, of the subject is measured at about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, or about 80 weeks.
In some embodiments, solanezumab is administered to the patient who is a carrier of at least one allele of APOE e4 and causes at least a 5% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of at least one allele of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of at least one allele of APOE s4 and causes at least a 15% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 20% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 25% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 60% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the MMSE scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the MMSE scale.
In some embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the CDS- SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 10% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 50% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 55% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the CDS-SB scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the CDS-SB scale.
In some embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADCS-ADL scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADCS- ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADCS- ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADCS-ADL scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADCS-ADL scale.
In some embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADCS-iADL scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADCS-iADLs scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADCS-iADLs scale.
In some embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 10% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 15% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 35% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 55% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 60% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 65% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the ADAS-Cogl4 scale.
In embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 5% slowing of cognitive decline versus placebo on the iADRS scale In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 10% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 15% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 20% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 25% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 30% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 35% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 40% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 45% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 50% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 55% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE s4 and causes at least a 60% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 65% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 70% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 75% slowing of cognitive decline versus placebo on the iADRS scale. In other embodiments, solanezumab is administered to the patient who is a carrier of APOE e4 and causes at least a 80% slowing of cognitive decline versus placebo on the iADRS scale.
In some embodiments, the antibody of the present disclosure can be administered in simultaneous, separate, or sequential combination with an effective amount of a symptomatic agent to treat Alzheimer’s disease. Symptomatic agents can be selected from cholinesterase inhibitors (ChEIs) and/or a partial /V-methyl-D-aspartate (NMDA) antagonists. In a preferred embodiment the agent is a ChEI. In another preferred embodiment the agent is a NMDA antagonist or a combination agent comprising a ChEI and NMDA antagonist.
In some embodiments, the disease characterized by Ab deposit in the brain of the subject is selected from preclinical Alzheimer’s disease, clinical AD, prodromal AD, mild AD, moderate AD, severe AD, Down’s syndrome, clinical cerebral amyloid angiopathy, or pre clinical cerebral amyloid angiopathy. In some embodiments, the subject is an early symptomatic AD patient. In some embodiments, the subject has prodromal AD and mild dementia due to AD. In some embodiments, the human subject has preclinical AD. In embodiments, the human subject has evidence of elevated brain amyloid.
In some embodiments, the subject has a genetic mutation that causes autosomal- dominant Alzheimer’s disease or at a higher risk for developing AD by virtue of carrying one or two APOE e4 alleles. In particular embodiments, the subject carries one or two APOE s4 alleles, /. e. , the patient is heterozygous or homozygous. The present disclosure includes use of biomarkers of a disease characterized by Ab deposits in the brain of a human subject, including Alzheimer’s disease. Such biomarkers include, e.g ., amyloid deposits, amyloid plaque, Ab in CSF, Ab in the plasma, brain tau deposition, tau in plasma, or tau in cerebrospinal fluid and their use in screening, diagnosis, treatment, or prevention. Non-limiting potential uses of such biomarkers include: 1) identification of subjects destined to become affected or who are in the “preclinical” stages of a disease; 2) reduction in disease heterogeneity in clinical trials or epidemiologic studies; 3) reflection of the natural history of disease encompassing the phases of induction, latency, and detection; and 4) target subjects for a clinical trial or for treatment/prevention of a disease.
In some embodiments, the biomarkers may be used to assess whether a subject can be treated using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers may be used to assess whether a disease (as described herein) can be prevented in the subject using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers can be used to assess whether a subject is responsive to treatment or prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers can be used to stratify or classify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of diseases (as described herein) using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the biomarkers may be used to assess disease state of a subject and/or the duration for administration of the antibodies or doses thereof, as described herein, to the subject.
In some embodiments, the subject has a genetic mutation that causes autosomal- dominant Alzheimer’s disease or at a higher risk for developing AD by virtue of carrying one or two APOE e4 alleles. In particular embodiments, the subject carries one or two APOE s4 alleles. Such patients, when administered solanezumab have better clinical outcomes than non-carriers.
In some embodiments, the subject has low to moderate tau burden or has been determined to have low to moderate tau burden. The subject has low to moderate tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18F-flortaucipir) is from <1.10 standardized uptake value ratio (SUVr) to <1.46 SUVr. In some embodiments, the subject has low to moderate tau burden or has been determined to have low to moderate tau burden and carries one or two APOE e4 alleles.
In some embodiments, the subject has very low tau burden or has been determined to have very low tau burden. The subj ect has very low tau burden if the tau burden as measured by PET brain imaging (using, e.g ., 18F-flortaucipir) is less than 1.10 SUVr. In some embodiments, the subject has very low tau burden or has been determined to have very low tau burden and carries one or two APOE e4 alleles.
In some embodiments, the subject has very low to moderate tau burden or has been determined to have very low tau to moderate tau burden. The subject has very low to moderate tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18F- flortaucipir) is <1.46 SUVr. In some embodiments, the subject has very low to moderate tau burden or has been determined to have very low to moderate tau burden and carries one or two APOE e4 alleles.
In some embodiments, the subject does not have a high tau burden or has been determined to not have a high tau burden. In some embodiments, the human subject has high tau burden if the tau burden as measured by PET brain imaging (using, e.g. , 18F- flortaucipir) is greater than 1.46 SUVr. In some embodiments, a subject with high tau is not administered the antibodies of the present disclosure. In some embodiments, the subject has does not have high tau burden or has been determined to not have a high tau burden and carries one or two APOE e4 alleles.
In some embodiments, solanezumab, the dosing regimen, or the methods described the present disclosure is efficacious in human subjects having very low to moderate tau. In some embodiments, solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having low to moderate tau. In some embodiments, solanezumab is most efficacious in human subjects having a tau level i) less than or equal to about 1.14 SUVr or ii) from about 1.14 SUVr to about 1.27 SUVr.
In some embodiments, solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having very low to moderate tau and carrying one or two APOE e4 alleles. In some embodiments, solanezumab, the dosing regimen, or the method described the present disclosure is efficacious in human subjects having low to moderate tau and carrying one or two APOE e4 alleles. In some embodiments, solanezumab is most efficacious in human subjects.
The tau level of a human subject can be determined by techniques and methods familiar to the diagnosing physician or a person of ordinary skill in the art. In some embodiments, a human subject, who is suffering from a disease characterized by amyloid beta (Ab) deposits, is determined to have very low to moderate tau, low to moderate tau, or no high tau using techniques and methods familiar to the diagnosing physician or a person of ordinary skill in the art. In some embodiments, such methods can also be used to prescreen, screen, diagnose, evaluate increase or reduction in brain tau burden, and/or to assess the progress achieved in the treatment or prevention of the diseases described herein. In some embodiments, the methods can also be used to stratify subjects into groups and/or to identify which group of subjects is responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein. In some embodiments, the methods or techniques used to determine/detect tau level of a human subject can be used for prescreening or screening subjects and determining which subjects are responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
For the purposes of the present disclosure, the tau level of a human subject can be determined using techniques or methods that, e.g ., detect or quantitate i) brain tau deposition, ii) tau in plasma, or iii) tau in cerebrospinal fluid. In some embodiments, brain tau burden, tau in plasma, or tau in cerebrospinal fluid can be used to stratify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of diseases (described herein) using the antibodies, the dosing regimen, or the methods described herein.
Tau levels in the brain of human subject can be determined using methods, such as, tau imaging with radiolabeled PET compounds (Leuzy et al ., “Diagnostic Performance of R0948 F18 Tau Positron Emission Tomography in the Differentiation of Alzheimer Disease from Other Neurodegenerative Disorders,” JAMA Neurology 77.8:955-965 (2020); Ossenkoppele et al., “Discriminative Accuracy of 18F-flortaucipir Positron Emission Tomography for Alzheimer Disease vs Other Neurodegenerative Disorders,” JAAJA 320, 1151-1162, doi:10.1001/jama.2018.12917 (2018), which are hereby incorporated by reference in their entireties. In some embodiments, the biomarker 18F-flortaucipir, which is a PET ligand, may be used for the purposes of the present disclosure. PET tau images can be, for example, quantitatively evaluated to estimate an SUVr (standardized uptake value ratio) by published methods (Pontecorvo et al ., “A Multicentre Longitudinal Study of Flortaucipir (18F) in Normal Ageing, Mild Cognitive Impairment and Alzheimer's Disease Dementia,” Brain 142: 1723-35 (2019); Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” Journal of Nuclear Medicine 59:937-43 (2018); Southekal e/ a/., “Flortaucipir FI 8 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-51 (2018), which are hereby incorporated by reference in their entireties) and/or to visually evaluate patients, e.g ., to determine whether the patient has an AD pattern (Fleisher etal ., “Positron Emission Tomography Imaging With 18F -flortaucipir and Postmortem Assessment of Alzheimer Disease Neuropathologic Changes,” JAMA Neurology 77:829-39 (2020), which is hereby incorporated by reference in its entirety). Lower SUVr values indicate less tau burden while higher SUVr values indicate a higher tau burden. In an embodiment, quantitative assessment by a flortaucipir scan is accomplished through an automated image processing pipeline as described in Southekal et al., “Flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2018), which is hereby incorporated by reference in its entirety. In some embodiments, counts within a specific target region of interest in the brain (e.g, multiblock barycentric discriminant analysis or MUBADA, see Devous et al., “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med. 59:937-943 (2018), which is hereby incorporated by reference in its entirety) are compared with a reference region wherein the reference region is, e.g, whole cerebellum, (wholeCere), cerebellar GM (cereCrus), atlas-based white matter (atlasWM), subject- specific WM (ssWM, e.g, using parametric estimate of reference signal intensity (PERSI), see Southekal et al., “Flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2018), which is hereby incorporated by reference in its entirety). A preferred method of determining tau burden is a quantitative analysis reported as a standardized uptake value ratio (SUVr), which represents counts within a specific target region of interest in the brain (e.g, MUBADA,) when compared with a reference region (e.g, using PERSI). In some embodiments, phosphorylated tau (P-tau; either phosphorylated at threonine 181 or 217) can be used to measure the tau load/burden for the purposes of the present disclosure (Barthelemy et al, “Cerebrospinal Fluid Phospho-tau T217 Outperforms T181 as a Biomarker for the Differential Diagnosis of Alzheimer's Disease and PET Amyloid positive Patient Identification,” Alzheimer’s Res. Ther. 12, 26, doi: 10.1186/sl3195-020- 00596-4 (2020); Mattsson et al., “Ab Deposition is Associated with Increases in Soluble and Phosphorylated Tau that Precede a Positive Tau PET in Alzheimer’s Disease,” Science Advances 6, eaaz2387 (2020), which are hereby incorporated by reference their entireties). In a particular embodiment, antibodies directed against human tau phosphorylated at threonine at residue 217 can be used to measure the tau load/burden in a subject for the purposes of the present disclosure (see International Patent Application Publication No. WO 2020/242963, which is incorporated by reference in its entirety). The present disclosure includes, in some embodiments, the use of anti-tau antibodies disclosed in WO 2020/242963 to measure the tau load/burden in a subject. The anti-tau antibodies disclosed in WO 2020/242963 are directed against isoforms of human tau expressed in the CNS ( e.g ., recognizing the isoforms expressed in the CNS and not recognizing isoforms of human tau expressed exclusively outside the CNS). Such antibodies against isoforms of human tau expressed in the CNS can be used in a method of identifying/selecting a patient as one or more of: (i) having a disease disclosed herein; (ii) at risk for having a disease disclosed herein; (iii) in need of treatment for a disease disclosed herein; or (iv) in need of neurological imaging.
A subject is positive for amyloid deposits when amyloid is detected in the brain by methods such as, amyloid imaging with radiolabeled PET compounds or using a diagnostic that detects Ab or a biomarker for Ab. Exemplary methods that can be used in the present disclosure to measure the brain amyloid load/burden include, e.g., Florbetapir (Carpenter, et al., “The Use of the Exploratory IND in the Evaluation and Development of 18F-PET Radiopharmaceuticals for Amyloid Imaging in the Brain: A Review of One Company's Experience,” The Quarterly Journal of Nuclear Medicine and Molecular Imaging 53.4:387 (2009), which is hereby incorporated by reference in its entirety); Florbetaben (Syed et al, “[18F]Florbetaben: A Review in b-Amyloid PET Imaging in Cognitive Impairment,” CNS Drugs 29, 605-613 (2015), which is hereby incorporated by reference in its entirety); and Flutemetamol (Heurling et al, “Imaging b-amyloid Using [18F] Flutemetamol Positron Emission Tomography: From Dosimetry to Clinical Diagnosis,” European Journal of Nuclear Medicine and Molecular Imaging 43.2: 362-373 (2016), which is hereby incorporated by reference in its entirety).
18F-florbetapir can provide a qualitative and quantitative measurement of brain plaque load in patients, including patients with prodromal AD or mild AD dementia. For example, the absence of significant 18]-florbetapir signal on a visual read indicates patients clinically manifesting cognitive impairment have sparse to no amyloid plaques. As such, 18F- florbetapir also provides a confirmation of amyloid pathology. 18F-Florbetapir PET also provides quantitative assessment of fibrillar amyloid plaque in the brain and, in some embodiments, can be used to assess amyloid plaque reductions from the brain by antibodies of the present disclosure.
Amyloid imaging with radiolabeled PET compounds can also be used to determine if Ab deposit in the brain of a human patient is reduced or increased (e.g, to calculate the percentage reduction in Ab deposit post treatment or to assess the progression of AD). A person of skill in the art can correlate the standardized uptake value ratio (SUVr) values obtained from amyloid imaging (with radiolabeled PET compounds) to calculate the % reduction in Ab deposit in the brain of the patient before and after treatment. The SUVr values can be converted to standardized centiloid (CL) units, where 100 is average for AD and 0 is average for young controls, allowing comparability amongst amyloid PET tracers, and calculation of reduction according to centiloid units (Klunk et al ., “The Centiloid Project: Standardizing Quantitative Amyloid Plaque Estimation by PET,” Alzheimer ’s & Dementia 11.1: 1-15 (2015) and Navitsky etal ., “Standardization of Amyloid Quantitation with Florbetapir Standardized Uptake Value Ratios to the Centiloid Scale,” Alzheimer's & Dementia 14.12: 1565-1571 (2018), which are hereby incorporated by reference in their entireties). In some embodiments, the change in brain amyloid plaque deposition from baseline is measured by 18F-florbetapir PET scan.
Cerebrospinal fluid or plasma-based analysis of b-amyloid can also be used to measure the amyloid load/burden for the purposes of the present disclosure. For example, Ab42 can be used to measure brain amyloid (Palmqvist, S. et al ., “Accuracy of Brain Amyloid Detection in Clinical Practice Using Cerebrospinal Fluid Beta-amyloid 42: a Cross- validation Study Against Amyloid Positron Emission Tomography. JAMA Neurol 71, 1282-1289 (2014), which is hereby incorporated by reference in its entirety). In some embodiments, the ratio of Ab42/Ab40 or Ab42/Ab38 can be used as a biomarker for amyloid beta (Janelidze etal ., “CSF Abeta42/Abeta40 and Abeta42/Abeta38 Ratios: Better Diagnostic Markers of Alzheimer Disease,” Ann Clin Transl Neurol 3, 154-165 (2016), which is hereby incorporated by reference in its entirety).
In some embodiments, deposited brain amyloid plaque or Ab in CSF or plasma can be used to stratify subjects into groups and to identify which group of subjects is responsive to treatment/prevention of a disease (as described herein) using the antibodies, the dosing regimen, or the methods described herein.
In some embodiment, solanezumab is administered by intravenous infusion. In another embodiment, solanezumab is administered subcutaneously.
Solanezumab binds selectively to Ab found in the brain. Exemplary embodiments of anti-Ab antibodies of the present disclosure include solanezumab, which is described in (including methods of making and using it) in the following patent documents, which are expressly incorporated herein by reference: US Patent No. 7,195,761, US Patent Application Publication No. 20060039906, US Patent No. 7,892,545, US Patent No. 8,591,894, US Patent No, 7,771,722, US Patent Application Publication No. 20070190046.
Skilled artisans will recognize that solanezumab is an IgGl monoclonal antibody having complementarity-determining regions (CDRs). Solanezumab binds to the mid domain of the Ab peptide. In some embodiments, the antibody has the following sequence.
Light Chain (SEQ ID NO: 1):
D VVMTQ SPL SLP VTLGQP ASISCRS SQ SLIY SDGN AYLHWFLQKPGQ SPRLLIYK V SNRF S GVPDRF S GS GS GTDF TLKI SRVE AED VGV Y Y C S Q S THVP WTF GQGTK VEI KRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ E S VTEQD SKD STYSLSSTLTL SK AD YEKHK V Y ACE VTHQGL S SP VTK SFNRGEC
Heavy Chain (SEQ ID NO: 2):
EVQLVESGGGLVQPGGSLRLSCAASGFTFSRYSMSWVRQAPGKGLELVAQINSV GNSTYYPDTVKGRFTISRDNAKNTLYLQMNSLRAEDTAVYYCASGDYWGQGTL VTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFP AVLQ S SGL Y SL S SWT VP S S SLGTQT YICNVNHKP SNTKVDKKVEPK SCDK THT CPPCP APELLGGP S VFLFPPKPKDTLMISRTPEVT C VVYD V SHEDPEVKFNW Y VDGVEVHNAKTKPREEQYNSTYRVV S VLTVLHQDWLNGKEYKCKV SNKALPAP IERTISRARGQPREPQVYTLPPSRDELTRNQVSLTCLVRGFYPSDIAVEWESNGQP ENN YRTTPP VLD SDGSFFL Y SKLTVDKSRWQQGNVF SC S VMHEALHNH YT QKSL SLSPGK
The antibody of the preceding sequence (as well the antibodies of the patents noted above in the following patent documents US Patent No. 7,195,761, US Patent Application Publication No. 20060039906, US Patent No. 7,892,545, US Patent No. 8,591,894, US Patent No, 7,771,722, US Patent Application Publication No. 20070190046) may be formulated and referred to as solanezumab. Thus, the present embodiments include antibodies with SEQ ID NOs: 1 and 2 or other antibodies, as well as those antibodies which have been formulated in a composition called solanezumab. Those skilled in the art will appreciate that any of the antibodies recited in the present disclosure may be used.
Information about Solanezumab used in combination with other antibodies is found in US Patent Application Publication No. 20190382471, which is expressly incorporated herein by reference in its entirety.
As used herein, an “antibody” is an immunoglobulin molecule comprising two HC and two LC interconnected by disulfide bonds. The amino terminal portion of each LC and HC includes a variable region responsible for antigen recognition via the complementarity determining regions (CDRs) contained therein. The CDRs are interspersed with regions that are more conserved, termed framework regions. Assignment of amino acids to CDR domains within the LCVR and HCVR regions of the antibodies of the present disclosure is based on the following: Rabat numbering convention (Rabat, et al ., Ann. NY Acad. Sci. 190:382-93 (1971); Rabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 (1991)), and North numbering convention (North et al., A New Clustering of Antibody CDR Loop Conformations, Journal of Molecular Biology, 406:228-256 (2011)). Following the above method, the CDRs of the antibodies of the present disclosure were determined.
The antibodies of the present disclosure are monoclonal antibodies (“mAbs”). Monoclonal antibodies can be produced, for example, by hybridoma technologies, recombinant technologies, phage display technologies, synthetic technologies, e.g., CDR- grafting, or combinations of such or other technologies known in the art. The monoclonal antibodies of the present disclosure are human or humanized. Humanized antibodies can be engineered to contain one or more human framework regions (or substantially human framework regions) surrounding CDRs derived from a non-human antibody. Human framework germline sequences can be obtained from ImunoGeneTics (INGT) via their website (imgt.cines.fr), or from The Immunoglobulin FactsBook by Marie-Paule Lefranc and Gerard Lefranc, Academic 25 Press, 2001, ISBN 01244135 l.Techinques for generating human or humanized antibodies are well known in the art. In another embodiment of the present disclosure, the antibody, or the nucleic acid encoding the same, is provided in isolated form. As used herein, the term “isolated” refers to a protein, peptide or nucleic acid that is not found in nature and is free or substantially free from other macromolecular species found in a cellular environment. “Substantially free,” as used herein, means the protein, peptide or nucleic acid of interest comprises more than 80% (on a molar basis) of the macromolecular species present, preferably more than 90% and more preferably more than 95%.
Solanezumab may be administered as a pharmaceutical composition. The pharmaceutical composition comprising an antibody of the present disclosure can be administered to a subject at risk for, or exhibiting, diseases or disorders as described herein by parental routes ( e.g. , subcutaneous, intravenous, intraperitoneal, intramuscular). Subcutaneous and intravenous routes are preferred.
The terms “treatment,” “treating” or “to treat” and the like include restraining, slowing, or stopping the progression or severity of an existing symptom, condition, disease, or disorder in a subject. The term “subject” refers to a human subject or a patient.
The term “prevention” means prophylactic administration of the antibody of the present disclosure to an asymptomatic subject or a subject with pre-clinical Alzheimer’s disease to prevent onset or progression of the disease.
The terms “disease characterized by deposition of Ab” or a “disease characterized by Ab deposits” are used interchangeably and refer to a disease that is pathologically characterized by Ab deposits in the brain or in brain vasculature. This includes diseases such as Alzheimer’s disease, Down’s syndrome, and cerebral amyloid angiopathy. A clinical diagnosis, staging or progression of Alzheimer’s disease can be readily determined by the attending diagnostician or health care professional, as one skilled in the art, by using known techniques and by observing results. This generally includes brain plaque imaging, mental or cognitive assessment (e.g, Clinical Dementia Rating - summary of boxes (CDR- SB), Mini -Mental State Exam (MMSE) or Alzheimer’s Disease Assessment Scale- Cognitive (ADAS-Cog)) or functional assessment (e.g, Alzheimer’s Disease Cooperative Study-Activities of Daily Living (ADCS-ADL); Alzheimer's Disease Cooperative Study - instrumental items of the Activities of Daily Living Inventory (ADCS-iADL)). The cognitive and functional assessment can be used to determine changes in a patient’s cognition (e.g, cognitive decline) and function (e.g, functional decline). “Clinical Alzheimer’s disease” as used herein is a diagnosed stage of Alzheimer’s disease. It includes conditions diagnosed as prodromal Alzheimer’s disease, mild Alzheimer’s disease, moderate Alzheimer’s disease, and severe Alzheimer’s disease. The term “pre-clinical Alzheimer’s disease” is a stage that precedes clinical Alzheimer’s disease, where measurable changes in biomarkers (such as CSF Ab42 levels or deposited brain plaque by amyloid PET) indicate the earliest signs of a patient with Alzheimer’s pathology, progressing to clinical Alzheimer’s disease. This is usually before symptoms such as memory loss and confusion are noticeable. Pre-clinical Alzheimer’s disease also includes pre-symptomatic autosomal dominant carriers, as well as patients with higher risk for developing AD by virtue of carrying one or two APOE s4 alleles.
A reduction or slowing of cognitive decline can be measured by cognitive assessments such as Clinical Dementia Rating - summary of boxes (CDR-SB), Mini-Mental State Exam (MMSE), or Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-Cog). A reduction or slowing of functional decline can be measured by functional assessments such as Alzheimer’s Disease Competence Scale- Activities of Daily Living (ADCS-ADL).
As used herein, “mg/kg” means an amount, in milligrams, of antibody or drug administered to a subject based on his or her bodyweight in kilograms. A dose is given at one time. For example, a 10 mg/kg dose of antibody for a subject weighing 70 kg would be a single 700 mg dose of antibody given in a single administration. Similarly, a 20 mg/kg dose of antibody for a subject weighing 70 kg would be a 1400 mg dose of antibody given at a single administration.
As used herein, a human subject has “very low tau” burden if the tau burden is less than 1.10 SUVr (<1.10 SUVr) using 18F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (multiblock barycentric discriminant analysis or MUBADA, see Devous el al., “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18,” ./. Nucl. Med. 59:937-943 (2018)) when compared with a reference region (parametric estimate of reference signal intensity or PERSI, see , e.g. , Southekal et al ., “Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2018)). See also, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
As used herein, a human subject has “very low tau to moderate tau” burden if the tau burden is less than or equal to 1.46 SUVr (i.e., <1.46 SUVr) using 18F -flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18,” J. Nucl. Med. 59:937-943 (2018)) when compared with a reference region (PERSI, see , Southekal et al. , “Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2018)). See also, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
As used herein, a human subject has “low tau to moderate tau” burden if the tau burden is from greater than or equal to 1.10 to less than or equal to 1.46 (i.e., <1.10 SUVr to <1.46 SUVr) using 18F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med. 59:937-943 (2018)) when compared with a reference region (PERSI, see , Southekal et al. , “Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J. Nucl. Med. 59:944-951 (2018)). “Low tau to moderate tau” burden can also be referred to as “intermediate” tau burden. See also, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
As used herein, a human subject has “high tau” burden if the tau burden is greater than 1.46 SUVr (i.e., >1.46 SUVr) using 18F-flortaucipir based quantitative analysis where quantitative analysis refers to calculation of SUVr and SUVr represents counts within a specific target region of interest in the brain (MUBADA, see Devous et al. , “Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir FI 8,” J. Nucl. Med. 59:937- 943 (2018)) when compared with a reference region (PERSI, see, Southekal et al., “Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity,” J Nucl. Med. 59:944-951 (2018)). See also, International Patent Application No. PCT/US2022/011894, which is hereby incorporated by reference in its entirety.
As used herein, the term “about” means up to ±10%.
The terms “human subject” and “patient” are used interchangeably in the present disclosure.
As used herein, “methods of treatment” are equally applicable to use of a composition for treating the diseases or disorders described herein and/or compositions for use and/or uses in the manufacture of a medicaments for treating the diseases or disorders described herein.
EXAMPLES
The following Figures further illustrate the present disclosure. It should be understood however, that the Examples are set forth by way of illustration and not limitation, and that various modifications may be made by one of ordinary skill in the art.
Example 1: Clinical Trials for Solanezumab
Expedition-1 and Expedition-2: Alzheimer’s patients were studied in two phase 3, double-blind trials (Expedition- 1 and Expedition-2, ClinicalTrials.gov numbers, NCT00905372 andNCT00904683, respectively), which randomly assigned 1012 and 1040 patients, respectively, with mild-to-moderate Alzheimer's disease to receive placebo or solanezumab (administered intravenously at a dose of 400 mg) every 4 weeks for 18 months. (Doody, etal. , “Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer’s Disease”, New England Journal of Medicine, 370; 4, pp. 311-321 (2014), which is hereby incorporated by reference in its entirety). The primary outcomes were the changes from baseline to week 80 in scores on the 11 -item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cogl l; range, 0 to 70, with higher scores indicating greater cognitive impairment) and the Alzheimer's Disease Cooperative Study- Activities of Daily Living scale (ADCS-ADL; range, 0 to 78, with lower scores indicating worse functioning). After analysis of data from Expedition- 1, the primary outcome for Expedition-2 was revised to the change in scores on the 14-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cogl4; range, 0 to 90, with higher scores indicating greater impairment), in patients with mild Alzheimer's disease.
Both trials involved otherwise healthy patients 55 years of age or older who had mild- to-moderate Alzheimer's disease without depression. Mild-to-moderate Alzheimer's disease was documented on the basis of a score of 16 to 26 on the Mini-Mental State Examination (MMSE; score range, 0 to 30, with higher scores indicating better cognitive function) and the criteria of the National Institute of Neurological and Communicative Disorders and Stroke- Alzheimer's Disease and Related Disorders Association. The absence of depression was documented on the basis of a score of 6 or less on the Geriatric Depression Scale (score range, 0 to 15, with higher scores indicating more severe depression). Participants were randomly assigned to receive solanezumab (400 mg) or placebo, administered as an intravenous infusion of approximately 70 ml over a period of 30 minutes, once every 4 weeks for 18 months. Concomitant treatment with cholinesterase inhibitors, memantine, or both was allowed.
Expedition-3 : A double-blind, placebo-controlled, phase 3 trial involving patients with mild dementia due to Alzheimer’s disease, defined as a Mini-Mental State Examination (MMSE) score of 20 to 26 (on a scale from 0 to 30, with higher scores indicating better cognition) and with amyloid deposition shown by means of florbetapir positron-emission tomography or Ab1-42 measurements in cerebrospinal fluid (Expedition-3, ClinicalTrials.gov number, NCT01900665). Patients were randomly assigned to receive solanezumab at a dose of 400 mg or placebo intravenously every 4 weeks for 76 weeks. The primary outcome was the change from baseline to week 80 in the score on the 14-item cognitive subscale of the Alzheimer’s Disease Assessment Scale (ADAS-Cogl4; scores range from 0 to 90, with higher scores indicating greater cognitive impairment). (Honig, et al., “Trial of Solanezumab for Mild Dementia Due to Alzheimer’s Disease,” New England Journal of Medicine, vol 78, No. 4, pp. 321-300 (2018), which is hereby incorporated by reference in its entirety). This trial included male and female patients, 55 to 90 years of age, who met the diagnostic criteria for probable Alzheimer’s disease according to the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association. Unlike the Expedition- 1 and Expedition-2 trials, the Expedition-3 trial included only patients with mild Alzheimer’s disease who had biomarker evidence of amyloid-related disease, determined by means of either florbetapir positron-emission tomography (PET) scan or Ab1-42 measurements in cerebrospinal fluid (CSF).
Patients were randomly assigned in double-blind fashion to receive intravenous infusions of either solanezumab at a dose of 400 mg or placebo every 4 weeks for 76 weeks. Patients who completed the double-blind period could participate in an optional 24-month open-label period. Concomitant therapy, including treatments for symptoms of dementia (acetylcholinesterase inhibitors and memantine, alone or in combination) and nondrug treatments, was allowed in order to ensure that patients continued receiving the standard of care for Alzheimer’s disease. The primary objective of the trial was to test the hypothesis that solanezumab would slow the cognitive decline of Alzheimer’s disease, as compared with placebo, in patients with mild dementia due to Alzheimer’s disease.
Analysis of data from Expedition- 1, Expedition-2 and Expedition-3 show that solanezumab shows better clinical outcomes in carriers of APOE e4 than non-carriers. The better clinical outcomes are shown on the Integrated AD Rating Scale (iADRS; a primary endpoint which is a composite tool measuring cognition and daily function), the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB), and ADAS-Cog scale. A 42% slowing in decline on the ADAS-Cogl4 scale was observed in Expedition-1 and Expedition-2 trials. In Expedition-3 in Mild AD Dementia patients, there was a 15% slowing in decline.
Table 2 shows that administration of solanezumab to APOE e4 carriers of Expedition- 3 performed better in slowing decline than non-carriers:
Table 2: Change in MMSE based upon APOE e4 status.
Figure imgf000050_0001
Table 3: Change in Expedition-3 of CDS-SB by APOE e4 status.
Figure imgf000050_0002
Table 4: Change in ADCS-ADL by APOE e4 status in Expedition-3.
Figure imgf000050_0003
Figure imgf000051_0001
Thus, as can be seen from this data, when a human subject who is a carrier of APOE e4 is given solanzumab, that subject performs better on clincial endpoints. In Table 2, the APOE s4 carriers had a -19% slowing of cognitive decline vs. placebo on the MMSE scale (Figures 9 and 10A-B). In Table 3, the carriers had a 18% slowing of cognitive decline vs. placebo on the CDR-SB scale (Figures 11 and 12A-B). In Table 4, the carriers had a 17% slowing of cognitive decline vs. placebo on the ADCS-ADL scale (Figures 13 and 14A-B). Additionally, carriers had a 17% slowing of cognitive decline vs. placebo in Expedition-3 and a 28% slowing in pooled data from Expedition- 1 and -2 on the ADCS-iADL scale. The APOE e4 carriers had a 15% slowing of cognitive decline vs. placebo in
Expedition-3 (Figures 4A, 4B) and a 42% slowing in cognitive decline in pooled data from Expedition-1 and -2 (Figures 5A and 5B).
Figure 15 shows a change on the iADRS scale between the carriers and non-carriers of APOE s4. Specifically, the carriers had a 11% slowing of cognitive decline vs. placebo in Expedition-3.
In Expedition-3, for treatment with low-dose of solanezumab (400mg IV every month), all outcome measures had a larger effect size in the APOE e4 carrier group relative to the non-carrier group although treatment differences did not reach significance for the iADRS and ADAS-Cogl4. In the CDR-SB, solanezumab reduced the clinical decline by 18% (p=0.009) at 80 weeks. In the ADCS-iADL and the MMSE, the slowing rates were respectively 17% (p=0.017) and 19% (p=0.005), and the treatment differences were significant as early as 64 weeks for both scales. Effect sizes were larger in all clinical outcome measures in pooled mild AD dementia data from Expedition-1 and Expedition-2. Low-dose solanezumab slowed the clinical decline by 42% as measured with the ADAS- Cogl4 (p<0.001) and by 28% in the ADCS-iADL (p<0.01) in APOE e4 carriers. In the 3 clinical studies (Expedition- 1, -2, and -3), solanezumab did not affect the clinical state of APOE e4 non-carriers. Outcome measures show higher effect sizes for carriers compared to non-carriers and often larger and significant treatment differences in the carrier population. Across all therapeutic clinical trials selected for the presence of amyloid pathology, at baseline, carriers are younger and have higher amyloid loads and higher tau pathology. The clinical decline across all scales for the placebo groups does not differ by carrier status. For placebo groups, comparison of carrier status shows there is no significant longitudinal change in amyloid, however there is a trend toward greater tau change in carriers vs. non carriers. The relative longitudinal change of amyloid while on therapy shows a greater decrease in non-carriers than carriers. One hypothesis to consider is the interaction of APOE s4 with tau. It has been known that amyloid deposition concentrates and contains APOE s4 embedded within the plaque. More recently, it was shown that APOE e4 is also isolated with tau tangles. Animal data further suggest that there is an APOE e4 interaction with tau. In addition, a rare mutation in APOE e4 appeared to protect a subject, carrying an autosomal dominant PSEN1 mutation, well beyond the age of typical onset despite having substantial brain amyloid load but relatively low tau burden. Data suggests that APOE e4 also influences tau beyond an amyloid interaction and that the rate of tau change may be faster in carriers. Furthermore, the impact of treatment may have a greater influence on tau progression which is more directly linked to clinical progression. Tau progression and spread has been linked to LRP 1. (Rauch, etal., “LRP l is a Master Regulator of T au Uptake and Spread,” Nature ; 580(7803):381-385 (2020), which is hereby incorporated by reference in its entirety). Recently reported, LRP1 appears to facilitate tau internalization and degradation via an APOE e4 mediated mechanism. ( See Cooper el al ., “Regulation of Tau Internalization, Degradation, and Seeding by LRP1 Reveals Multiple Pathways for Tau Catabolism,” Journal of Biological Chemistry , Vol. 296 (2021) (which is hereby incorporated by reference in its entirety). So, LRP1 seems ideally linked given its interaction with APOE e4 and tau and the interaction of APOE e4 with amyloid and tau.

Claims

WE CLAIM:
1. A method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having an APOE e4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subject that has one or two alleles of APOE e4.
2. A method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
3. A method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE e4 allele; and ii) if the human subject is identified as having an APOE e4 allele, administering or having administered a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
4. A method of treating or preventing a disease characterized by amyloid beta (Ab) deposits in the brain of a human subject, comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE s4 allele.
5. A method according to any of claims 1-4, wherein the antibody is solanezumab.
6. A method according to any of claim 1-5 wherein said step of administering comprises administering one or more than one dose of the antibody.
7. A method according to any of claims 1 -6 wherein the human subj ect has two APOE s4 alleles.
8. The method according to any one of claims 1-7, wherein the disease characterized by Ab deposit in the brain of the human subject is selected from preclinical Alzheimer’s disease (AD), clinical AD, prodromal AD, mild AD, moderate AD, severe AD, Down’s syndrome, clinical cerebral amyloid angiopathy, or pre-clinical cerebral amyloid angiopathy.
9. The method according to any one of claims 1-7, wherein the human subject is administered the antibody for a duration sufficient to treat or prevent the disease.
10. The method according to any one of claims 1 -9, wherein the treatment or prevention of the disease slows cognitive or functional decline in the human subject.
11. The method according to any one of claims 1 -9, wherein the treatment or prevention of the disease reduces amyloid load in the brain of the human subject.
12. The method according to any one of claims 1-11, the antibody is administered to the subject till amyloid deposits or plaques are cleared from the subject’s brain.
13. The method according to claim 11, wherein the reduction in amyloid load in the brain of the human subject is determined by amyloid PET brain imaging or a diagnostic that detects a biomarker for Ab.
14. The method according to claim 1, wherein the human subject has early symptomatic AD.
15. The method according to claim 14, wherein the human subject has prodromal AD and/or mild dementia due to AD.
16. The method according to claim 1, wherein the human subject has preclinical AD.
17. The method according to claim 16, wherein the human subject has evidence of elevated brain amyloid.
18. The method according to any one of claims 1-17, wherein the antibody is administered at a dose of 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, 1600 mg every 4 weeks, 2000 mg every 4 weeks, 2400 mg every 4 weeks, 2800 mg every 4 weeks, 3200 mg every 4 weeks, 3600 mg every 4 weeks, 4000 mg every 4 weeks, 4400 mg every 4 weeks, 4800 mg every 4 weeks, 5200 mg every 4 weeks, or 5600 mg every 4 weeks.
19. The method of claim 18, wherein the antibody is administered at an initial dose of 400 mg every 4 weeks and is titrated to 1600 mg every 4 weeks.
20. The method of claim 18, wherein the antibody is administered at an initial dose of 1600 mg every 4 weeks and is titrated to 400 mg every 4 weeks.
21. A method of reducing amyloid load in the brain of a human subj ect having an APOE s4 allele comprising: i) administering a therapeutically effective amount of an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2, or pharmaceutical composition thereof, to the human subject that has one or two alleles of APOE e4.
22. A method of reducing amyloid load in the brain of a human subject having been identified as having an APOE e4 allele, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
23. A method of reducing amyloid load in the brain of a human subject, comprising: i) identifying or having identified the human subject as having an APOE e4 allele; and ii) if the human subject is identified as having an APOE e4 allele, administering or having administered an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject.
24. A method of reducing amyloid load in the brain of a human subject, comprising: i) administering an antibody comprising SEQ ID NO: 1 and SEQ ID NO: 2 to the human subject, wherein the human subject has an APOE e4 allele.
25. A method according to any of claims 21-24, wherein the antibody is solanezumab.
26. A method according to any of claim 21-25 wherein said step of administering comprises administering one or more than one dose of the antibody.
27. A method according to any of claims 21-26 wherein the human subject has two APOE s4 alleles.
28. The method according to any one of claims 21-27, wherein the disease characterized by Ab deposit in the brain of the human subject is selected from preclinical Alzheimer’s disease (AD), clinical AD, prodromal AD, mild AD, moderate AD, severe AD, Down’s syndrome, clinical cerebral amyloid angiopathy, or pre-clinical cerebral amyloid angiopathy.
29. The method according to any one of claims 21-27, wherein the human subject is administered the antibody for a duration sufficient to treat or prevent the disease.
30. The method according to any one of claims 21-29, wherein the treatment or prevention of the disease slows cognitive or functional decline in the human subject.
31. The method according to any one of claims 21-30, the antibody is administered to the subject till amyloid deposits or plaques are cleared from the subject’s brain.
32. The method according to claim 31, wherein the reduction in amyloid load in the brain of the human subject is determined by amyloid PET brain imaging or a diagnostic that detects a biomarker for Ab.
33. The method according to claim 21, wherein the human subject has early symptomatic AD.
34. The method according to claim 33, wherein the human subject has prodromal AD and/or mild dementia due to AD.
35. The method according to claim 21, wherein the human subject has preclinical AD.
36. The method according to claim 35, wherein the human subject has evidence of elevated brain amyloid.
37. The method according to any one of claims 21-36, wherein the antibody is administered at a dose of 400 mg every 4 weeks, 800 mg every 4 weeks, 1200 mg every 4 weeks, 1600 mg every 4 weeks, 2000 mg every 4 weeks, 2400 mg every 4 weeks, 2800 mg every 4 weeks, 3200 mg every 4 weeks, 3600 mg every 4 weeks, 4000 mg every 4 weeks, 4400 mg every 4 weeks, 4800 mg every 4 weeks, 5200 mg every 4 weeks, or 5600 mg every 4 weeks.
38. The method of claim 37, wherein the antibody is administered at an initial dose of 400 mg every 4 weeks and is titrated to 1600 mg every 4 weeks.
39. The method of claim 38, wherein the antibody is administered at an initial dose of 1600 mg every 4 weeks and is titrated to 400 mg every 4 weeks.
40. An antibody for use in the treatment or prevention of a disease characterized by Ab deposits in the brain of a human subject, wherein the human subject carries one or two alleles of APOE e4 and wherein the antibody comprises a light chain of SEQ ID NO: 1 and a heavy chain of SEQ ID NO: 2.
41. The antibody for use according to claim 40, wherein the disease characterized by Ab deposit in the brain of the human subject is selected from preclinical Alzheimer’s disease (AD), clinical AD, prodromal AD, mild AD, moderate AD, severe AD, Down’s syndrome, clinical cerebral amyloid angiopathy, or pre-clinical cerebral amyloid angiopathy.
42. The antibody for use according to any one of claims 40-41, wherein the human subject is an early symptomatic AD patient or wherein the human subject has prodromal AD and mild dementia due to AD.
PCT/US2022/030167 2021-05-24 2022-05-20 Anti-amyloid beta antibodies and uses thereof WO2022251048A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163192291P 2021-05-24 2021-05-24
US63/192,291 2021-05-24

Publications (1)

Publication Number Publication Date
WO2022251048A1 true WO2022251048A1 (en) 2022-12-01

Family

ID=82156813

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/030167 WO2022251048A1 (en) 2021-05-24 2022-05-20 Anti-amyloid beta antibodies and uses thereof

Country Status (1)

Country Link
WO (1) WO2022251048A1 (en)

Citations (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
US6815175B2 (en) 2001-03-16 2004-11-09 Cornell Research Foundation, Inc. Anti-amyloid peptide antibody based diagnosis and treatment of a neurological disease or disorder
US20060039906A1 (en) 2000-02-24 2006-02-23 Holtzman David M Humanized antibodies that sequester abeta peptide
US7060270B2 (en) 2001-11-02 2006-06-13 Diagenics International Corporation Methods and compositions of monoclonal antibodies specific for beta-amyloid proteins
US7122374B1 (en) 2002-04-09 2006-10-17 Takaomi Saido Amyloid beta-protein 3(pE)-42 antibodies and uses thereof
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7189819B2 (en) 2000-12-06 2007-03-13 Wyeth Humanized antibodies that recognize beta amyloid peptide
US7256273B2 (en) 2002-03-12 2007-08-14 Elan Pharma International Limited Humanized antibodies that recognize beta amyloid peptide
US20070190046A1 (en) 2004-02-23 2007-08-16 Eli Lilly And Company Anti-abeta antibody
US7318923B2 (en) 2001-04-30 2008-01-15 Eli Lilly And Company Humanized anti-βantibodies
US7320790B2 (en) 2001-04-30 2008-01-22 Eli Lilly And Company Humanized antibodies
US7339035B2 (en) 2001-08-03 2008-03-04 Medical & Biological Laboratories Co., Ltd. Antibody recognizing GM1 ganglioside-bound amyloid β-protein and DNA encoding the antibody
US7575880B1 (en) 2000-05-26 2009-08-18 Elan Pharma International Limited Method of screening an antibody for activity in clearing an amyloid deposit
US7625560B2 (en) 2004-12-15 2009-12-01 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US7731962B2 (en) 2005-02-14 2010-06-08 Merck & Co., Inc. Anti-ADDL monoclonal antibody and use thereof
US7741448B2 (en) 2005-06-21 2010-06-22 Medical & Biological Laboratories Co., Ltd. Antibody having inhibitory effect on amyloid fibril formation
US7763249B2 (en) 2004-04-27 2010-07-27 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Human anti-amyloid β peptide antibody and fragment of said antibody
US7763250B2 (en) 2005-04-29 2010-07-27 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and nucleic acids encoding same
US7771722B2 (en) 2001-08-17 2010-08-10 Eli Lilly And Company Assay method for alzheimer's disease
US7772375B2 (en) 2005-12-12 2010-08-10 Ac Immune S.A. Monoclonal antibodies that recognize epitopes of amyloid-beta
US7780963B2 (en) 2004-10-25 2010-08-24 Merck & Co., Inc. Anti-ADDL antibodies and uses thereof
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7807165B2 (en) 2004-07-30 2010-10-05 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US7807157B2 (en) 2004-02-20 2010-10-05 Intellect Neurosciences Inc. Monoclonal antibodies and use thereof
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7892544B2 (en) 2006-07-14 2011-02-22 Ac Immune Sa Humanized anti-beta-amyloid antibody
US7902328B2 (en) 2003-01-31 2011-03-08 Abbott Laboratories Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US7906625B2 (en) 2005-01-24 2011-03-15 Amgen Inc. Humanized anti-amyloid antibody
US7932048B2 (en) 2005-03-09 2011-04-26 Consejo Superior De Investigaciones Cientificas Method for the in vitro diagnosis of alzheimer's disease using a monoclonal antibody
US7939075B2 (en) 2007-01-11 2011-05-10 Philipps-Universitaet Marburg Human monoclonal anti-amyloid-beta antibodies
US7955812B2 (en) 2004-07-19 2011-06-07 The General Hospital Corporation Methods of diagnosing alzheimer's disease by detecting antibodies to cross-linked β-amyloid oligomers
US8025878B2 (en) 2006-03-23 2011-09-27 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US8105594B2 (en) 1998-05-21 2012-01-31 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US8106164B2 (en) 2004-06-21 2012-01-31 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
US8124076B2 (en) 2005-08-18 2012-02-28 Ramot At Tel Aviv University Ltd. Single chain antibodies against β-amyloid peptide
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US8227576B2 (en) 2006-03-30 2012-07-24 Glaxo Group Limited Antibodies against amyloid-β peptide
US8268973B2 (en) 2007-12-28 2012-09-18 Onclave Therapeutics Anti-amyloid antibodies
US8303954B2 (en) 2009-08-07 2012-11-06 Kyowa Hakko Kirin Co., Ltd Anti-Aβ oligomer humanized antibody
US8323654B2 (en) 2007-05-14 2012-12-04 Medtronic, Inc. Anti-amyloid beta antibodies conjugated to sialic acid-containing molecules
US8333967B2 (en) 2009-08-07 2012-12-18 Kyowa Hakko Kirin Co., Ltd Anti-Aβ oligomer humanized antibody
US8337848B2 (en) 2007-10-25 2012-12-25 University College Cardiff Consultants Limited Monoclonal antibody for APP
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
US8420093B2 (en) 2005-02-14 2013-04-16 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and use thereof
US8470321B2 (en) 2007-11-16 2013-06-25 The Rockefeller University Antibodies specific for the protofibril form of beta-amyloid protein
US8507206B2 (en) 2004-07-02 2013-08-13 Northwestern University Monoclonal antibodies that target pathological assemblies of amyloid β (Abeta)
US8614299B2 (en) 2009-05-12 2013-12-24 Sanofi Humanized antibodies specific to the protofibrillar form of the beta-amyloid peptide
US8710193B2 (en) 2009-10-16 2014-04-29 Kyoto University Antibody recognizing turn structure in amyloid β
US8795664B2 (en) 2010-06-04 2014-08-05 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin Monoclonal antibodies targeting amyloid beta oligomers
US8906370B2 (en) 2005-12-12 2014-12-09 Hoffmann-La Roche Inc. Antibodies against amyloid beta 4 with glycosylation in the variable region
US8906367B2 (en) 2007-01-05 2014-12-09 University Of Zurich Method of providing disease-specific binding molecules and targets
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US9051364B2 (en) 2008-11-19 2015-06-09 Forschungszentrum Juelich Gmbh Composition for producing anti-amyloid beta peptide antibodies with D-peptides
US9062102B2 (en) 2011-03-08 2015-06-23 Alzinova Ag Anti oligomer antibodies and uses thereof
US9125846B2 (en) 2010-10-15 2015-09-08 The Board Of Regents Of The University Of Texas System Antibodies that bind amyloid oligomers
US9133267B2 (en) 2005-11-22 2015-09-15 The Trustees Of The University Of Pennsylvania Antibody treatment of Alzheimer's and related diseases
US9146244B2 (en) 2007-06-12 2015-09-29 Ac Immune S.A. Polynucleotides encoding an anti-beta-amyloid monoclonal antibody
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US9309309B2 (en) 2010-07-14 2016-04-12 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and uses thereof
US9309307B2 (en) 2011-01-07 2016-04-12 Seiko Epson Corporation Antibody against amyloid precursor protein signal peptide
US9329189B2 (en) 2002-09-27 2016-05-03 Janssen Pharmaceutica Nv N-11 truncated amyloid-beta Aβ11-x monoclonal antibodies and compositions
US9573994B2 (en) 2014-07-10 2017-02-21 Bioarctic Neuroscience Ab Aβ protofibril binding antibodies
US9738712B2 (en) 2013-11-13 2017-08-22 F. Hoffman-La Roche Ag Camelid single-domain antibody directed against amyloid beta and methods for producing conjugates thereof
US9834598B2 (en) 2012-10-15 2017-12-05 Medimmune Limited Antibodies to amyloid beta
US9944696B2 (en) 2016-01-15 2018-04-17 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US10035847B2 (en) 2013-10-02 2018-07-31 The Rockefeller University Amyloid protofibril antibodies and methods of use thereof
US10112987B2 (en) 2012-01-09 2018-10-30 Icb International, Inc. Blood-brain barrier permeable peptide compositions comprising a vab domain of a camelid single domain heavy chain antibody against an amyloid-beta peptide
US10494425B2 (en) 2015-02-24 2019-12-03 Rpeptide, Llc Anti-amyloid-beta antibodies
US20190382471A1 (en) 2016-07-01 2019-12-19 Eli Lilly And Company ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
US10519223B2 (en) 2016-11-03 2019-12-31 Jannsen Pharmaceutica Nv Antibodies to pyroglutamate amyloid-β and uses thereof
US10603367B2 (en) 2015-07-16 2020-03-31 Probiodrug Ag Humanized antibodies
US10647759B2 (en) 2017-04-20 2020-05-12 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US10654917B2 (en) 2015-01-29 2020-05-19 Technophage, Investigacao E Desenvolvimento Em Biotecnologia, Sa Antibody molecules and peptide delivery systems for use in alzheimer's disease and related disorders
US10738109B2 (en) 2012-01-09 2020-08-11 Icb International, Inc. Blood-brain barrier permeable peptide compositions comprising a VAB domain of an anti-amyloid-beta camelid single-domain heavy-chain only antibody
WO2020242963A1 (en) 2019-05-31 2020-12-03 Eli Lilly And Company Compounds and methods targeting human tau

Patent Citations (111)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US8105594B2 (en) 1998-05-21 2012-01-31 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US7582733B2 (en) 1998-11-30 2009-09-01 Elan Pharma International Limited Humanized antibodies that recognize beta amyloid peptide
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
US7892545B2 (en) 2000-02-24 2011-02-22 Eli Lilly And Company Humanized antibodies that sequester amyloid beta peptide
US20060039906A1 (en) 2000-02-24 2006-02-23 Holtzman David M Humanized antibodies that sequester abeta peptide
US7195761B2 (en) 2000-02-24 2007-03-27 Eli Lilly And Company Humanized antibodies that sequester abeta peptide
US8591894B2 (en) 2000-02-24 2013-11-26 Eli Lilly And Company Humanized antibodies that sequester amyloid beta peptide
US7575880B1 (en) 2000-05-26 2009-08-18 Elan Pharma International Limited Method of screening an antibody for activity in clearing an amyloid deposit
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US7189819B2 (en) 2000-12-06 2007-03-13 Wyeth Humanized antibodies that recognize beta amyloid peptide
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6815175B2 (en) 2001-03-16 2004-11-09 Cornell Research Foundation, Inc. Anti-amyloid peptide antibody based diagnosis and treatment of a neurological disease or disorder
US7320790B2 (en) 2001-04-30 2008-01-22 Eli Lilly And Company Humanized antibodies
US7318923B2 (en) 2001-04-30 2008-01-15 Eli Lilly And Company Humanized anti-βantibodies
US7339035B2 (en) 2001-08-03 2008-03-04 Medical & Biological Laboratories Co., Ltd. Antibody recognizing GM1 ganglioside-bound amyloid β-protein and DNA encoding the antibody
US7771722B2 (en) 2001-08-17 2010-08-10 Eli Lilly And Company Assay method for alzheimer's disease
US7060270B2 (en) 2001-11-02 2006-06-13 Diagenics International Corporation Methods and compositions of monoclonal antibodies specific for beta-amyloid proteins
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
US7256273B2 (en) 2002-03-12 2007-08-14 Elan Pharma International Limited Humanized antibodies that recognize beta amyloid peptide
US7122374B1 (en) 2002-04-09 2006-10-17 Takaomi Saido Amyloid beta-protein 3(pE)-42 antibodies and uses thereof
US9329189B2 (en) 2002-09-27 2016-05-03 Janssen Pharmaceutica Nv N-11 truncated amyloid-beta Aβ11-x monoclonal antibodies and compositions
US9939452B2 (en) 2002-09-27 2018-04-10 Janssen Pharmaceutica Nv N-11 truncated amyloid-beta monoclonal antibodies, compositions, methods and uses
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US7902328B2 (en) 2003-01-31 2011-03-08 Abbott Laboratories Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7807157B2 (en) 2004-02-20 2010-10-05 Intellect Neurosciences Inc. Monoclonal antibodies and use thereof
US20070190046A1 (en) 2004-02-23 2007-08-16 Eli Lilly And Company Anti-abeta antibody
US8222002B2 (en) 2004-04-27 2012-07-17 Juridical Foundation The Chemo-Sero-Therapeuric Research Institute Human anti-amyloid beta peptide antibody and fragment of said antibody
US7763249B2 (en) 2004-04-27 2010-07-27 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Human anti-amyloid β peptide antibody and fragment of said antibody
US8106164B2 (en) 2004-06-21 2012-01-31 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
US8507206B2 (en) 2004-07-02 2013-08-13 Northwestern University Monoclonal antibodies that target pathological assemblies of amyloid β (Abeta)
US7955812B2 (en) 2004-07-19 2011-06-07 The General Hospital Corporation Methods of diagnosing alzheimer's disease by detecting antibodies to cross-linked β-amyloid oligomers
US8425905B2 (en) 2004-07-30 2013-04-23 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US7807165B2 (en) 2004-07-30 2010-10-05 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US8268593B2 (en) 2004-07-30 2012-09-18 Rinat Neuroscience Corp. Polynucleotides encoding antibodies directed against amyloid-beta peptide
US7927594B2 (en) 2004-07-30 2011-04-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide
US8383113B2 (en) 2004-10-25 2013-02-26 Northwestern University Anti-ADDL antibodies and uses thereof
US7780963B2 (en) 2004-10-25 2010-08-24 Merck & Co., Inc. Anti-ADDL antibodies and uses thereof
US7811563B2 (en) 2004-10-25 2010-10-12 Northwestern University Anti-addl antibodies and uses thereof
US8889138B2 (en) 2004-10-25 2014-11-18 Merck Sharp & Dohme Corp. Anti-ADDL antibodies and uses thereof
US8128930B2 (en) 2004-10-25 2012-03-06 Northwestern University Anti-ADDL antibodies and uses thereof
US7625560B2 (en) 2004-12-15 2009-12-01 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US7906625B2 (en) 2005-01-24 2011-03-15 Amgen Inc. Humanized anti-amyloid antibody
US7731962B2 (en) 2005-02-14 2010-06-08 Merck & Co., Inc. Anti-ADDL monoclonal antibody and use thereof
US8420093B2 (en) 2005-02-14 2013-04-16 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and use thereof
US8105593B2 (en) 2005-02-14 2012-01-31 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and use thereof
US7932048B2 (en) 2005-03-09 2011-04-26 Consejo Superior De Investigaciones Cientificas Method for the in vitro diagnosis of alzheimer's disease using a monoclonal antibody
US8398978B2 (en) 2005-04-29 2013-03-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US7763250B2 (en) 2005-04-29 2010-07-27 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and nucleic acids encoding same
US7741448B2 (en) 2005-06-21 2010-06-22 Medical & Biological Laboratories Co., Ltd. Antibody having inhibitory effect on amyloid fibril formation
US8124076B2 (en) 2005-08-18 2012-02-28 Ramot At Tel Aviv University Ltd. Single chain antibodies against β-amyloid peptide
US8221750B2 (en) 2005-08-18 2012-07-17 Ramot At Tel-Aviv University Ltd. Single chain antibodies against β-amyloid peptide
US9133267B2 (en) 2005-11-22 2015-09-15 The Trustees Of The University Of Pennsylvania Antibody treatment of Alzheimer's and related diseases
US7772375B2 (en) 2005-12-12 2010-08-10 Ac Immune S.A. Monoclonal antibodies that recognize epitopes of amyloid-beta
US8906370B2 (en) 2005-12-12 2014-12-09 Hoffmann-La Roche Inc. Antibodies against amyloid beta 4 with glycosylation in the variable region
US9272031B2 (en) 2005-12-12 2016-03-01 F. Hoffmann-La Roche Inc. Antibodies against amyloid beta 4 with glycosylation in the variable region
US8025878B2 (en) 2006-03-23 2011-09-27 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US8227576B2 (en) 2006-03-30 2012-07-24 Glaxo Group Limited Antibodies against amyloid-β peptide
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
US8246954B2 (en) 2006-07-14 2012-08-21 Ac Immune S.A. Methods of treating amyloidosis with humanized anti-beta-amyloid antibodies
US7892544B2 (en) 2006-07-14 2011-02-22 Ac Immune Sa Humanized anti-beta-amyloid antibody
US8124353B2 (en) 2006-07-14 2012-02-28 Ac Immune S.A. Methods of treating and monitoring disease with antibodies
US8906367B2 (en) 2007-01-05 2014-12-09 University Of Zurich Method of providing disease-specific binding molecules and targets
US7939075B2 (en) 2007-01-11 2011-05-10 Philipps-Universitaet Marburg Human monoclonal anti-amyloid-beta antibodies
US8491903B2 (en) 2007-01-11 2013-07-23 Philipps-Universitaet Marburg Method of treatment of neurodementing diseases using isolated, monoclonal, human, anti-B-amyloid antibody
US8323654B2 (en) 2007-05-14 2012-12-04 Medtronic, Inc. Anti-amyloid beta antibodies conjugated to sialic acid-containing molecules
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US9585956B2 (en) 2007-06-12 2017-03-07 Ac Immune S.A. Polynucleotides encoding anti-amyloid beta monoclonal antibodies
US9146244B2 (en) 2007-06-12 2015-09-29 Ac Immune S.A. Polynucleotides encoding an anti-beta-amyloid monoclonal antibody
US8337848B2 (en) 2007-10-25 2012-12-25 University College Cardiff Consultants Limited Monoclonal antibody for APP
US8470321B2 (en) 2007-11-16 2013-06-25 The Rockefeller University Antibodies specific for the protofibril form of beta-amyloid protein
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
US8268973B2 (en) 2007-12-28 2012-09-18 Onclave Therapeutics Anti-amyloid antibodies
US8636981B2 (en) 2007-12-28 2014-01-28 Onclave Therapeutics Detection of amyloid deposits using anti-amyloid antibodies
US9051364B2 (en) 2008-11-19 2015-06-09 Forschungszentrum Juelich Gmbh Composition for producing anti-amyloid beta peptide antibodies with D-peptides
US9382312B2 (en) 2009-05-12 2016-07-05 Sanofi Humanized antibodies specific to the protofibrillar form of the beta-amyloid peptide
US8614299B2 (en) 2009-05-12 2013-12-24 Sanofi Humanized antibodies specific to the protofibrillar form of the beta-amyloid peptide
US10112991B2 (en) 2009-05-12 2018-10-30 Sanofi Humanized antibodies specific to the protofibrillar form of the beta-amyloid peptide
US8333967B2 (en) 2009-08-07 2012-12-18 Kyowa Hakko Kirin Co., Ltd Anti-Aβ oligomer humanized antibody
US8303954B2 (en) 2009-08-07 2012-11-06 Kyowa Hakko Kirin Co., Ltd Anti-Aβ oligomer humanized antibody
US9181332B2 (en) 2009-08-07 2015-11-10 Kyowa Hakko Kirin Co., Ltd Anti-A(β) oligomer humanized antibody
US8710193B2 (en) 2009-10-16 2014-04-29 Kyoto University Antibody recognizing turn structure in amyloid β
US8795664B2 (en) 2010-06-04 2014-08-05 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin Monoclonal antibodies targeting amyloid beta oligomers
US9309309B2 (en) 2010-07-14 2016-04-12 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and uses thereof
US9125846B2 (en) 2010-10-15 2015-09-08 The Board Of Regents Of The University Of Texas System Antibodies that bind amyloid oligomers
US9895429B2 (en) 2010-10-15 2018-02-20 The Board Of Regents Of The University Of Texas System Antibodies that bind amyloid oligomers
US9309307B2 (en) 2011-01-07 2016-04-12 Seiko Epson Corporation Antibody against amyloid precursor protein signal peptide
US9062102B2 (en) 2011-03-08 2015-06-23 Alzinova Ag Anti oligomer antibodies and uses thereof
US10738109B2 (en) 2012-01-09 2020-08-11 Icb International, Inc. Blood-brain barrier permeable peptide compositions comprising a VAB domain of an anti-amyloid-beta camelid single-domain heavy-chain only antibody
US10112987B2 (en) 2012-01-09 2018-10-30 Icb International, Inc. Blood-brain barrier permeable peptide compositions comprising a vab domain of a camelid single domain heavy chain antibody against an amyloid-beta peptide
US9834598B2 (en) 2012-10-15 2017-12-05 Medimmune Limited Antibodies to amyloid beta
US10662239B2 (en) 2012-10-15 2020-05-26 Medimmune Limited Antibodies to amyloid beta
US10035847B2 (en) 2013-10-02 2018-07-31 The Rockefeller University Amyloid protofibril antibodies and methods of use thereof
US9738712B2 (en) 2013-11-13 2017-08-22 F. Hoffman-La Roche Ag Camelid single-domain antibody directed against amyloid beta and methods for producing conjugates thereof
US9573994B2 (en) 2014-07-10 2017-02-21 Bioarctic Neuroscience Ab Aβ protofibril binding antibodies
US10654917B2 (en) 2015-01-29 2020-05-19 Technophage, Investigacao E Desenvolvimento Em Biotecnologia, Sa Antibody molecules and peptide delivery systems for use in alzheimer's disease and related disorders
US10494425B2 (en) 2015-02-24 2019-12-03 Rpeptide, Llc Anti-amyloid-beta antibodies
US10603367B2 (en) 2015-07-16 2020-03-31 Probiodrug Ag Humanized antibodies
US9944696B2 (en) 2016-01-15 2018-04-17 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US20190382471A1 (en) 2016-07-01 2019-12-19 Eli Lilly And Company ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
US10519223B2 (en) 2016-11-03 2019-12-31 Jannsen Pharmaceutica Nv Antibodies to pyroglutamate amyloid-β and uses thereof
US10851156B2 (en) 2016-11-03 2020-12-01 Janssen Pharmaceutica Nv Methods of detecting pyroglutamate amyloid beta protein (3pE Aβ) using anti-3pE Aβ antibodies
US10647759B2 (en) 2017-04-20 2020-05-12 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
WO2020242963A1 (en) 2019-05-31 2020-12-03 Eli Lilly And Company Compounds and methods targeting human tau

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
AISEN ET AL.: "The Future of Anti-amyloid Trials", THE JOURNAL OF PREVENTION OF ALZHEIMER'S DISEASE, vol. 7, 2020, pages 146 - 151
BARTHELEMY ET AL.: "Cerebrospinal Fluid Phospho-tau T217 Outperforms T181 as a Biomarker for the Differential Diagnosis of Alzheimer's Disease and PET Amyloid-positive Patient Identification", ALZHEIMER'S RES. THER., vol. 12, 2020, pages 26
BRASHEAR ET AL.: "Clinical Evaluation of Amyloid-related Imaging Abnormalities in Bapineuzumab Phase III Studies", J. OF ALZHEIMER'S DISEASE, vol. 66, no. 4, 2018, pages 1409 - 1424
BUDD ET AL.: "Clinical Development of Aducanumab, an Anti-Ap Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer's Disease", THE JOURNAL OF PREVENTION OF ALZHEIMER'S DISEASE, vol. 4, no. 4, 2017, pages 255, XP055768746, DOI: 10.14283/jpad.2017.39
CARLSON CHRISTOPHER ET AL: "Amyloid-related imaging abnormalities from trials of solanezumab for Alzheimer's disease", ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING, vol. 2, no. 1, 1 March 2016 (2016-03-01), pages 75 - 85, XP055954467, ISSN: 2352-8729, DOI: 10.1016/j.dadm.2016.02.004 *
CARPENTER ET AL.: "The Use of the Exploratory IND in the Evaluation and Development of F-PET Radiopharmaceuticals for Amyloid Imaging in the Brain: A Review of One Company's Experience", THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 53, no. 4, 2009, pages 387
COOPER ET AL.: "Regulation of Tau Internalization, Degradation, and Seeding by LRP1 Reveals Multiple Pathways for Tau Catabolism", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 296, 2021
DEVOUS ET AL.: "Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F 18", J. NUCL. MED., vol. 59, 2018, pages 937 - 943
DEVOUS ET AL.: "Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18", J. NUCL. MED., vol. 59, 2018, pages 937 - 943
DEVOUS ET AL.: "Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F18", JOURNAL OF NUCLEAR MEDICINE, vol. 59, 2018, pages 937 - 43
DOODY ET AL., NEW ENGLAND JOURNAL OF MEDICINE, vol. 370, no. 4, 2014, pages 311 - 321
DOODY ET AL.: "Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer's Disease", NEW ENGLAND JOURNAL OF MEDICINE, vol. 370, no. 4, 2014, pages 311 - 321, XP055183832, DOI: 10.1056/NEJMoa1312889
DORAISWAMY ET AL.: "Amyloid-P Assessed by Florbetapir F18 PET and 18-month Cognitive Decline: A Multicenter Study", NEUROLOGY, vol. 79, 2012, pages 1636 - 44
EARLEY LAURIEL F. ET AL: "Adeno-associated Virus (AAV) Assembly-Activating Protein Is Not an Essential Requirement for Capsid Assembly of AAV Serotypes 4, 5, and 11", JOURNAL OF VIROLOGY, vol. 91, no. 3, 1 February 2017 (2017-02-01), US, XP055953369, ISSN: 0022-538X, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5244341/pdf/e01980-16.pdf> DOI: 10.1128/JVI.01980-16 *
FARLOW MARNOLD SEVAN DYCK CH ET AL.: "Safety and biomarker effects of solanezumab in patients with Alzheimer's disease", ALZHEIMER'S DEMENT, vol. 8, 2012, pages 261 - 71
FARLOW MARTIN ET AL: "Safety and biomarker effects of solanezumab in patients with Alzheimer's disease", ALZHEIMER'S & DEMENTIA, vol. 8, no. 4, 7 June 2012 (2012-06-07), US, pages 261 - 271, XP055923662, ISSN: 1552-5260, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1016/j.jalz.2011.09.224> DOI: 10.1016/j.jalz.2011.09.224 *
FLEISHER ADAM S ET AL: "Use of white matter reference regions for detection of change in florbetapir positron emission tomography from completed phase 3 solanezumab trials", ALZHEIMER'S & DEMENTIA, vol. 13, no. 10, 10 October 2017 (2017-10-10), pages 1117 - 1124, XP085225601, ISSN: 1552-5260, DOI: 10.1016/J.JALZ.2017.02.009 *
FLEISHER ET AL.: "Associations Between Biomarkers and Age in the Presenilin 1 E280A Autosomal Dominant Alzheimer Disease Kindred: A Cross-sectional Study", JAMA NEUROL, vol. 72, 2015, pages 316 - 24
FLEISHER ET AL.: "Positron Emission Tomography Imaging With F-flortaucipir and Postmortem Assessment of Alzheimer Disease Neuropathologic Changes", JAMA NEUROLOGY, vol. 77, 2020, pages 829 - 39
HARDY ET AL.: "The Amyloid Hypothesis of Alzheimer's Disease: Progress and Problems on the Road to Therapeutics", SCIENCE, vol. 297, 2002, pages 353 - 6, XP055310378, DOI: 10.1126/science.1072994
HEURLING ET AL.: "Imaging 0-amyloid Using e8F] Flutemetamol Positron Emission Tomography: From Dosimetry to Clinical Diagnosis", EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 43, no. 2, 2016, pages 362 - 373, XP035871023, DOI: 10.1007/s00259-015-3208-1
HONIG ET AL.: "Trial of Solanezumab for Mild Dementia Due to Alzheimer's Disease", NEW ENGLAND JOURNAL OF MEDICINE, vol. 78, no. 4, 2018, pages 321 - 300
JANELIDZE ET AL.: "CSF Abeta42/Abeta40 and Abeta42/Abeta38 Ratios: Better Diagnostic Markers of Alzheimer Disease", ANN CLIN TRANSL NEUROL, vol. 3, 2016, pages 154 - 165
JONSSON ET AL.: "A Mutation in APP Protects Against Alzheimer's Disease and Age-related Cognitive Decline", NATURE, vol. 488, 2012, pages 96 - 9, XP055675990, DOI: 10.1038/nature11283
KABAT ET AL., ANN. NY ACAD. SCI., vol. 190, 1971, pages 382 - 93
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
KEVIN M. SCHILLING ET AL: "Paper and toner three-dimensional fluidic devices: programming fluid flow to improve point-of-care diagnostics", LAB ON A CHIP, vol. 13, no. 4, 1 January 2013 (2013-01-01), UK, pages 628, XP055283832, ISSN: 1473-0197, DOI: 10.1039/c2lc40984d *
KLEIN ET AL.: "Gantenerumab Reduces Amyloid-P Plaques in Patients with Prodromal to Moderate Alzheimer's Disease: a PET Substudy Interim Analysis", ALZHEIMER'S RESEARCH & THERAPY, vol. 11, no. 1, 2019, pages 101
KLUNK ET AL.: "The Centiloid Project: Standardizing Quantitative Amyloid Plaque Estimation by PET", ALZHEIMER'S & DEMENTIA, vol. 11, no. 1, 2015, pages 1 - 15, XP055930663, DOI: 10.1016/j.jalz.2014.07.003
LEUZY ET AL.: "Diagnostic Performance of R0948 F18 Tau Positron Emission Tomography in the Differentiation of Alzheimer Disease from Other Neurodegenerative Disorders", JAMA NEUROLOGY, vol. 77, no. 8, 2020, pages 955 - 965
MARIE-PAULE LEFRANCGERARD LEFRANC: "The Immunoglobulin FactsBook", 2001, ACADEMIC 25 PRESS
MASTERS ET AL.: "Alzheimer's Disease", NAT. REV. DIS. PRIMERS, vol. 1, 2015, pages 15056
MATTSSON ET AL.: "Ap Deposition is Associated with Increases in Soluble and Phosphorylated Tau that Precede a Positive Tau PET in Alzheimer's Disease", SCIENCE ADVANCES, vol. 6, 2020, pages eaaz2387
NAVITSKY ET AL.: "Standardization of Amyloid Quantitation with Florbetapir Standardized Uptake Value Ratios to the Centiloid Scale", ALZHEIMER'S & DEMENTIA, vol. 14, no. 12, 2018, pages 1565 - 1571, XP085545873, DOI: 10.1016/j.jalz.2018.06.1353
NORTH ET AL.: "A New Clustering of Antibody CDR Loop Conformations", JOURNAL OF MOLECULAR BIOLOGY, vol. 406, 2011, pages 228 - 256, XP028129711, DOI: 10.1016/j.jmb.2010.10.030
OSSENKOPPELE ET AL.: "Discriminative Accuracy of F-flortaucipir Positron Emission Tomography for Alzheimer Disease vs Other Neurodegenerative Disorders", JAMA, vol. 320, 2018, pages 1151 - 1162
PALMQVIST, S. ET AL.: "Accuracy of Brain Amyloid Detection in Clinical Practice Using Cerebrospinal Fluid Beta-amyloid 42: a Cross-validation Study Against Amyloid Positron Emission Tomography", JAMA NEUROL, vol. 71, 2014, pages 1282 - 1289
PIAZZAWINBLAD: "Amyloid-Related Imaging Abnormalities (ARIA) in Immunotherapy Trials for Alzheimer's Disease: Need for Prognostic Biomarkers?", JOURNAL OF ALZHEIMER'S DISEASE, vol. 52, 2016, pages 417 - 420, XP009195107, DOI: 10.3233/JAD-160122
PONTECORVO ET AL.: "A Multicentre Longitudinal Study of Flortaucipir (18F) in Normal Ageing, Mild Cognitive Impairment and Alzheimer's Disease Dementia", BRAIN, vol. 142, 2019, pages 1723 - 35
RAUCH ET AL.: "LRP1 is a Master Regulator of Tau Uptake and Spread", NATURE, vol. 580, no. 7803, 2020, pages 381 - 385, XP037092393, DOI: 10.1038/s41586-020-2156-5
SELKOE ET AL.: "The Amyloid Hypothesis of Alzheimer's Disease at 25 years", EMBO MOL. MED., vol. 8, 2016, pages 595 - 608
SELKOE: "The Origins of Alzheimer Disease: A is for Amyloid", JAMA, vol. 283, 2000, pages 1615 - 7
SIEMERS E R ET AL: "Safety and changes in plasma and cerebrospinal fluid amyloid [beta] after a single administration of an amyloid [beta] monoclonal antibody in subjects with Alzheimer disease", CLINICAL NEUROPHARMACOLOGY, RAVEN PRESS, NEW YORK, NY, US, vol. 33, no. 2, 1 January 2010 (2010-01-01), pages 67 - 73, XP009145612, ISSN: 0362-5664 *
SIEMERS ERFRIEDRICH SDEAN RA ET AL.: "Safety and changes in plasma and cerebrospinal fluid amyloid β after a single administration of an amyloid β monoclonal antibody in subjects with Alzheimer disease", CLIN. NEUROPHARMACOL., vol. 33, 2010, pages 67 - 73, XP009145612
SOUTHEKAL ET AL.: "Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity", J. NUCL. MED., vol. 59, 2018, pages 944 - 951
SOUTHEKAL ET AL.: "Flortaucipir F18 Quantitation Using Parametric Estimation of Reference Signal Intensity", J. NUCL. MED., vol. 59, 2018, pages 944 - 951
SPERLING ET AL.: "Amyloid-related Imaging Abnormalities in Patients with Alzheimer's Disease Treated with Bapineuzumab: A Retrospective Analysis", THE LANCET NEUROLOGY, vol. 11, no. 3, 2012, pages 241 - 249
SYED ET AL.: "F]Florbetaben: A Review in P-Amyloid PET Imaging in Cognitive Impairment", CNS DRUGS, vol. 29, 2015, pages 605 - 613

Similar Documents

Publication Publication Date Title
Mruthinti et al. Autoimmunity in Alzheimer’s disease: increased levels of circulating IgGs binding Aβ and RAGE peptides
Delrieu et al. Amyloid beta peptide immunotherapy in Alzheimer disease
JP2022145965A (en) Methods for treating alzheimer&#39;s disease
JP2017537905A (en) Treatment of Alzheimer&#39;s disease
TW202019471A (en) Methods of treatment and prevention of alzheimer&#39;s disease
US20240010713A1 (en) ANTI-N3pGlu AMYLOID BETA ANTIBODIES AND USES THEREOF
Plascencia-Villa et al. Lessons from antiamyloid-β immunotherapies in Alzheimer's disease
Yang et al. New developments of clinical trial in immunotherapy for Alzheimer's disease
US20240150450A1 (en) Anti-amyloid beta antibodies and uses thereof
US20240158486A1 (en) ANTI-N3pGlu AMYLOID BETA ANTIBODIES AND USES THEREOF
WO2022251048A1 (en) Anti-amyloid beta antibodies and uses thereof
WO2023283650A1 (en) Biomarkers for alzheimer&#39;s disease treatment
TWI843040B (en) ANTI-N3pGlu AMYLOID BETA ANTIBODIES AND USES THEREOF
WO2024107683A1 (en) ANTI-N3pGlu AMYLOID BETA ANTIBODIES, DOSES, AND USES THEREOF
CN116981690A (en) Anti-amyloid beta antibodies and uses thereof
Hawkes et al. Clinical immunotherapy trials in Alzheimer’s disease
TW202330030A (en) Treatment of a demyelinating disease of the central nervous system (cns) with satralizumab
WO2023149970A1 (en) Methods of treatment using p-tau181 level
Grundman et al. Immunotherapy for Alzheimer’s Disease
WO2024054416A1 (en) Scyllo-inositol in combination with immunotherapeutics for the treatment of alzheimer&#39;s disease
WO2023245008A1 (en) Methods of delaying or preventing the onset of alzheimer&#39;s disease using crenezumab
CN116916957A (en) anti-N3 pGlu amyloid beta antibodies and uses thereof
Khanna et al. Aducanumab. Anti-beta-amyloid monoclonal antibody, Treatment of Alzheimer
WO2023114586A1 (en) Methods of using an anti-amyloid beta protofibril antibody and anti-tau antibody

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: 22732729

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22732729

Country of ref document: EP

Kind code of ref document: A1