BIOMOLECULES INVOLVED IN ALZHEIMER'S DISEASE

Abstract
The invention relates to panels of biomarkers including proteins phosphatase 1 regulatory subunit 14A and/or 2′,3′-cyclic-nucleotide 3′-phosphodiesterase and/or phosphorylated tau or fragments thereof and methods using thereof for diagnosing, staging, treating and assessing the response of a treatment for a neurocognitive disorder characterised by tau toxicity, in particular for Alzheimer's disease. The present invention shows that the biomarkers disclosed herein are elevated in the brain of subjects with an advanced stage of a neurocognitive disorder (Braak stage V/VI) and/or are regulated in the CSF of AD subjects in comparison to cognitively affected non-AD controls; and/or regulated in response to two casein kinase 1 delta inhibitors.
Description
FIELD OF THE INVENTION

The invention relates to sets of biomarkers and methods using thereof for diagnosing, staging, treating and assessing the response of a treatment for a neurocognitive disorder characterised by tau toxicity, in particular for Alzheimer's disease.


BACKGROUND OF THE INVENTION

Neurofibrillary tangles, composed of intracellular aggregates of tau protein, are a key neuropathological feature of Alzheimer's disease (AD) and other neurodegenerative diseases, collectively termed tauopathies. Tau is an intracellular microtubule-associated protein, with several unique characteristics such as natively unfolded conformation, thermo-stability, acid-stability, and capability of post-translational modifications.


Abnormally hyperphosphorylated tau is a key feature of human tauopathies. In AD pathogenesis, accumulation of the amyloid-μ peptide (Aβ) interacts with the signalling pathways that regulate the phosphorylation of tau. Hyperphosphorylation of tau disrupts its normal function in regulating axonal transport and leads to the accumulation of neurofibrillary tangles and toxic species of soluble tau.


Currently there is no cure for AD. Approved treatments are few and of limited efficacy, serving mostly to slow or delay progression.


Identification and development of new therapies for the treatment of AD and other tauopathies is also greatly affected by the lack of effective diagnostic, prognostic and predictive biomarkers and by the lack of new targets for the design of new therapies. Currently, AD can only be definitively diagnosed by brain biopsy or upon autopsy after a patient has died. Clearly, in clinical settings, brain biopsy is rarely performed and diagnosis is still primarily made based on the history of the symptoms and depends on a battery of neurological, psychometric and biochemical tests. These latter tests include assessment of ApoE e4 allele status and measurements of amyloid beta, tau and phospho-tau in cerebrospinal fluid.


These present methods, nevertheless, are still unsatisfactory not only for the early diagnosis of AD and other tauopathies, but also for predicting the progression of neurological disease which is important for recruitment of patients for clinical trials, for designing new therapies and for predicting the effectiveness of current and new therapies.


The ideal diagnostic biomarkers should have high specificity for disease versus non-disease and high sensitivity to distinguish between disease types and stages.


Prognostic biomarkers should reflect the intensity and severity of the pathological changes and predict their future course from a very early stage of the disease, before degeneration is observed, until advanced stages of the disease.


Pharmacodynamic biomarkers should give a reliable indication whether an administered therapy is efficacious based on the changes in the level of disease-related proteins in readily accessible body fluids such as blood, blood products including platelets, serum and most preferably plasma and CSF. It is also desirable that such pharmacodynamics biomarkers can provide guidance to clinicians when to stop treatment or switch to a different therapy.


New targets need to be efficacious, safe, meet clinical and commercial needs and, above all, be ‘druggable’. A ‘druggable’ target is accessible to the putative drug molecule, be that a small molecule or larger biologicals and upon binding, elicit a biological response which may be measured both in vitro and in vivo. In other words, its inhibition or activation will result in a therapeutic effect in a disease state.


Hence, there remains a need for proteins that may perform with superior sensitivity and/or specificity as biomarkers in the diagnosis, staging, prognostic monitoring and assessment of the effectiveness of treatments for patients with Alzheimer's disease and other tauopathies and may serve as new targets for the development of new therapies.


SUMMARY OF THE INVENTION

The present invention, therefore, provides novel biomarkers and phosphorylated amino acids of tau or fragments thereof for use in methods for diagnosing, staging, treating and assessing the response of a treatment for a neurocognitive disorder characterised by tau toxicity such as tauopathies, in particular for Alzheimer's disease. In addition, the present invention provides novel targets for the development of new therapies against tauopathies or for repurposing existing therapies not originally designed for the treatment of neurocognitive disorders such as tauopathies.


In a first aspect the present invention provides for a panel of biomarkers comprising:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • and/or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


In a second aspect the present invention provides for a panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/5416 or S422;


      wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


In a third aspect the present invention provides for a panel of biomarkers comprising one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof.


In one embodiment of the first aspect, the panel of biomarkers further comprises one or more, alternative two or more, proteins selected from Group A, which comprises Actin alpha cardiac muscle 1 comprising or having the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising or having the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising or having the amino acid sequence of SEQ ID NO:3, cAMP-dependent protein kinase type I-beta regulatory subunit comprising or having the amino acid sequence of SEQ ID NO:24, Catenin delta-1 comprising or having the amino acid sequence of SEQ ID NO:4, Centrosomal protein of 170 kDa comprising or having the amino acid sequence of SEQ ID NO:23, Clathrin light chain B comprising or having the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising or having the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising or having the amino acid sequence of SEQ ID NO:14, GMP reductase 1 comprising or having the amino acid sequence of SEQ ID NO:27, Guanine nucleotide-binding protein G(q) subunit alpha comprising or having the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising or having the amino acid sequence of SEQ ID NO:15, KxDL motif-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:28, Lambda-crystallin homolog comprising or having the amino acid sequence of SEQ ID NO:18, Myelin-associated oligodendrocyte basic protein comprising or having the amino acid sequence of SEQ ID NO:20, Neutral alpha-glucosidase AB comprising or having the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising or having the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:16, Protein KIAA1045 comprising or having the amino acid sequence of SEQ ID NO:25, Secernin-2 comprising or having the amino acid sequence of SEQ ID NO:8, Serum albumin comprising or having the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising or having the amino acid sequence of SEQ ID NO:9, Synaptoporin comprising or having the amino acid sequence of SEQ ID NO:22, Syntaphilin comprising or having the amino acid sequence of SEQ ID NO:10, Transmembrane protein 119 comprising or having the amino acid sequence of SEQ ID NO: 21 and Tubulin alpha chain-like 3 comprising or having the amino acid sequence of SEQ ID NO:26.


In another embodiment, the panel of biomarkers according to the first aspect of the invention further comprises one or more proteins selected from Groups B, C or D.


In one other embodiment, the panel of biomarkers according to the first aspects and its embodiments may further comprise one or more biomarkers as defined in the second and/or third aspect of the invention.


In a fourth aspect the invention provides for a method for diagnosing a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers of the panel in said sample with reference concentrations or amounts of said biomarkers.


In a fifth aspect the invention provides for method for staging a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining the stage of the neurocognitive disorder in said subject by comparing said concentration or amount of each of the biomarkers of the panel in said sample with reference concentrations or amounts of said biomarkers.


In a sixth aspect the invention provides for a method for assessing in a subject the likelihood of developing a neurocognitive disorder, the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject is likely to develop a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers of the biomarker panel in said sample with reference concentrations or amounts of said biomarkers.


In a seventh aspect the invention provides for a method for treating a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers;
    • d) administering to said subject a drug for treating the neurocognitive disorder.


Alternatively, the seventh aspect of the present invention may be formulated as a drug for use in the treatment of a neurocognitive disorder in a subject, wherein the subject is selected by the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers.


Alternatively, the seventh aspect of the present invention may be formulated as a use of a drug for the manufacture of a medicament for the treatment of a neurocognitive disorder in a subject, wherein the subject is selected by the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers.


In an eighth aspect the present invention provides for a method for assessing the response to a drug for treating a neurocognitive disorder in a subject, wherein the subject has been treated or is being treated with said drug, the method comprising:

    • a) assaying a sample obtained from a subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has responded or is responding to said drug by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers.


In one embodiment of the seventh and eighth aspects of the invention, the drug for treating a neurocognitive disorder is a kinase inhibitor. Preferably, the kinase inhibitor is selected from a tau kinase inhibitor or a casein kinase inhibitor, optionally a casein kinase 1 alpha, beta, gamma, delta or epsilon. More preferably the kinase inhibitor is a casein kinase 1 delta inhibitor selected from 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine; 2-amino-3-[(thiophen-2-yl)carbonyl]indolizine-1-carboxamide; 2-[3-(pyridin-4-yl)-1H-pyrazol-4-yl]-1,3-benzoxazole; 2-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; 2-amino-3-benzoylindolizine-1-carboxamide; 2-amino-1-[(4-fluorophenyl)carbonyl]-1H-indole-3-carboxamide; combinations thereof; or pharmaceutically acceptable salt or solvate thereof. Even more preferably, the casein kinase 1 delta inhibitor is selected from 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine; 2-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide.


In preferred embodiments of the fourth, fifth, sixth, seventh and eighth aspect of the invention, the neurocognitive disorder is characterised by tau toxicity. More preferably, the neurocognitive disorder is a tauopathy.


The tauopathy may be selected from the group of Alzheimer's disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy (PSP), Pick's disease, corticobasal degeneration, multisystem atrophy (MSA), neurobasal degeneration with iron accumulation, type 1 (Hallervorden-Spatz), argyrophilic grain dementia, Down's syndrome, diffuse neurofibrillary tangles with calcification, dementia pugilistica, Gerstmann-Straussler-Scheinker disease, myotonic dystrophy, Niemann-Pick disease type C, progressive subcortical gliosis, prion protein cerebral amyloid angiopathy, tangle only dementia, postencephalitic parkinsonism, subacute sclerosing panencephalitis, Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex, non-Guamanian motor neuron disease with neurofibrillary tangles/dementia, chronic traumatic encephalopathy, alpha-synucleinopathies, Parkinson's disease or combinations thereof.


Even more preferably the tauopathy is Alzheimer's disease.


In one embodiment of the seventh aspect of the invention, step d) further comprises administering an additional therapeutic agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof.


In one other embodiment of the eighth aspect of the invention the subject may have also been treated or may also being treated with an additional therapeutic agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof.


In another embodiment of the eighth aspect, preferably or alternatively, after step c), the method comprises administering an additional therapeutic agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof.


In embodiments of the fourth, fifth, sixth, seventh and eighth aspect of the invention the assaying step a) and/or the measuring step b) comprise:

    • i) contacting said sample with one or more binding agents to each of said biomarkers of the panel; or
    • ii) detecting in said sample autoantibodies specific to each of said biomarkers; or
    • iii) detecting in said sample by mass spectrometry each of said biomarkers of the panel or fragments thereof, optionally by previously labelling said sample with one or more isobaric reactive mass labels; or
    • iv) detecting in said sample by 2D gel electrophoresis each of said biomarkers of the panel; or
    • iv) any combinations of i), ii), iii) or iv).


Preferably, the assaying in step a) and/or the measuring is step b) comprise:

    • i) detecting one or more fragments of said biomarkers in the panel and/or
    • ii) detecting one or more phosphorylated amino acids on tau comprising or having the amino acid sequence of SEQ ID NO: 29 or one or more fragments thereof; wherein when the phosphorylated amino acid on tau to be detected is T181, at least one more phosphorylated amino acid on tau or one or more fragments thereof is detected.


More preferably, the sample is immobilised on a solid support.


In embodiments of the fourth, fifth, sixth, seventh and eighth aspect of the invention the sample is selected from the group of cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue (e.g. brain tissue) or combinations thereof. Preferably the sample is CSF or blood. Also preferably, the subject is a human subject.


In a ninth aspect the present invention provides for a kit comprising reagents for assaying and/or measuring in a sample the biomarkers of a panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof.


The reagents may comprise one or more binding agents which specifically bind to the biomarkers of the panels. Preferably, the one or more binding agents are primary antibodies, wherein each primary antibody specifically binds to:

    • i) a different biomarkers of the panel and/or
    • ii) one or more phosphorylated amino acids of tau comprising or having amino acid sequence of SEQ ID NO: 29 or fragments thereof.


The reagents may further comprise one or more secondary antibodies which specifically bind to said primary antibodies. Preferably the secondary antibodies are labelled.


In another embodiment of the ninth aspect and of its embodiments the sample is selected from the group of cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue (e.g. brain tissue) or combinations thereof.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Sequence of human protein phosphatase 1 regulatory subunit 14A. Amino acids flagged by symbol □ or a * indicate amino acids which are replaced by a different or a modified amino acid in an isoform or a variant of human protein phosphatase 1 regulatory subunit 14A, respectively.



FIG. 2. Sequence of human 2′,3′-cyclic-nucleotide 3′-phosphodiesterase. Amino acids flagged by symbol □ or a * indicate amino acids which are replaced by a different or a modified amino acid in an isoform or a variant of human 2′,3′-cyclic-nucleotide 3′-phosphodiesterase, respectively.



FIG. 3. Venn diagram for protein phosphatase 1 regulatory subunit 14A.



FIG. 4. Venn diagram for 2′,3′-cyclic-nucleotide 3′-phosphodiesterase.



FIG. 5. Levels (as sums of ions intensities of all non-modified peptides) of protein phosphatase 1 regulatory subunit 14A in (A) Human brain with mild (Braak 0-II) (n=3), moderate (Braak III/IV) or severe (Braak v/VI) tau pathology; (B) Mouse brain from the TMHT model of human tauopathy treated orally with vehicle alone or vehicle including 30 mg/kg of casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine (PS110) and 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (PS278-05); and (C) CSF from cognitively affected non-AD controls (CTL; n=3) and patients with biochemically diagnosed AD (AD; n=3).



FIG. 6. Levels (as sums of ions intensities of all non-modified peptides) of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase in (A) Human brain with mild (Braak 0-II) (n=3), moderate (Braak III/IV) (n=3) or severe (Braak v/VI) (n=3) tau pathology; (B) Mouse brain from the TMHT model of human tauopathy treated orally with vehicle alone (n=3) or vehicle including 30 mg/kg of casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine (PS110; n=3) and 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (PS278-05; n=3); and (C) cerebrospinal fluid from cognitively affected non-AD controls (CTL; n=3) and patients with biochemically diagnosed AD (AD; n=3).



FIG. 7. Experimental design of TMTcalibrator™ experiments to measure brain derived proteins in CSF of human control and biochemically diagnosed AD subjects.





DEFINITIONS

The term “biomarker(s)” includes all biologically relevant forms of the protein identified, including post-translational modifications. For example, the biomarker can be present in a glycosylated, phosphorylated, multimeric, fragmented or precursor form. A biomarker fragment may be naturally occurring or, for example, enzymatically generated and still retaining the biologically active function of the full protein. Fragments will typically be at least about 10 amino acids, usually at least about 50 amino acids in length, and can be as long as 300 amino acids in length or longer.


The term “canonical sequence” is used herein as to refer to the most prevalent sequence and/or the most similar sequence among orthologous species. In particular, unless otherwise specified, the canonical sequence refers herein to the human sequence.


The term “KEGG pathway” refers to a collection of manually drawn pathway maps representing molecular interactions and reaction networks for metabolism, genetic information processing, environmental information processing, cellular processes, organismal systems, human diseases and drug development. “KEGG pathways mapping” is the process to map molecular datasets, especially large-scale datasets in genomics, transcriptomics, proteomics, and metabolomics, to the KEGG pathway maps for biological interpretation of higher-level systemic functions; (http://www.genome.jp/kegg/pathway.html).


The term “concentration or amount” refers to the relative concentration or amount of biomarker in the sample, for example as determined by LC-MS/MS label free quantification approaches such as area under the curve and spectral counting.


The term “comparing” or “compare” or grammatical equivalents thereof, means measuring the relative concentration or amount of a biomarker in a sample relative to other samples (for example protein concentrations or amounts stored in proprietary or public database).


The term “reference concentration or amount” refers to, but it is not limited to, protein concentrations or amounts stored in proprietary or public databases. The “reference concentration or amount” may have been obtained from a large screening of patients, or by reference to a known or previously determined correlation between such a determination and clinical information in control patients. For example, the reference values may be determined by comparison to the concentration or amount of the biomarkers in a control subject, for example a healthy person (i.e. without dementia) of similar age and gender as the subject. Alternatively, the reference values are values which can be found in literature such as the ApoE ε4 allele presence whereby the presence or absence of the mutations at position 112 and 158 represent the reference to be compared to, or like the levels of total tau (T-tau) >350 ng/L, phospho-tau (P-tau) >80 ng/L and Aβ42 <530 ng/L in the CSF (Hansson 0, et al., Lancet Neurol. 2006, 5(3), 228-34). In addition, the reference values may have been obtained from the same subject at one or more time points which precede in time the test time point. Such earlier sample may be taken one week or more, one month or more, three months or more, most preferably six months or more before the date of the test time point. In some embodiments, multiple earlier samples may be compared in a longitudinal manner and the slope of change in biomarker expression may be calculated as a correlate of cognitive decline.


The term “control” or as used herein “non AD control” or “non AD subject” refers to a tissue sample or a bodily fluid sample taken from a human or non-human subject that is cognitively normal or diagnosed with or presenting symptoms of a cognitive abnormality but defined, with respect to the existing biochemical tests, as non AD subjects.


The term “binding agent” generally refers to any molecule that has affinity for a biomarker of the present invention. Binding agents may comprises apatamers, antibodies, lectins and enzymes.


The term “antibody” includes polyclonal antiserum, monoclonal antibodies, fragments of antibodies such as single chain and Fab fragments, and genetically engineered antibodies. The antibodies may be chimeric or of a single species.


The term “aptamer” includes small affinity agents with selectivity for a specific target and which are polymers of nucleic acids, amino acids or other synthetic organic molecules or combinations of any of these constituent molecules.


The terms “selected reaction monitoring”, “SRM” and “MRM” means a mass spectrometry assay whereby precursor ions of known mass-to-charge ratio representing known biomarkers are preferentially targeted for analysis by tandem mass spectrometry in an ion trap or triple quadrupole mass spectrometer. During the analysis the parent ion is fragmented and the number of daughter ions of a second predefined mass-to-charge ratio is counted. Typically, an equivalent precursor ion bearing a predefined number of stable isotope substitutions but otherwise chemically identical to the target ion is included in the method to act as a quantitative internal standard.


The term “immunoassay” refers to any method of quantitatively measuring the level of one or more biomarkers of the present invention by capturing and or detecting the presence of the target biomarker using one or more binding agents. Immunoassays may be direct, where the biomarker is adsorbed onto a surface and detected using a binding agent carrying a detectable label, indirect where the biomarker is adsorbed onto a surface, a first binding agent with specificity for the biomarker is then captured onto the surface through specific binding to the target biomarker and detected using a second binding agent carrying a detectable label that is specific for the first antibody. Alternatively the immunoassay may be a sandwich immunoassay in which a binding agent is immobilised on a solid support and captures the target biomarker from the analytical sample. Subsequently, the now immobilised biomarker is detected using binding agents in the direct or indirect methods as described above.


The term “bead suspension array” means an immunoassay where the biomarker is detected on one or mall solid particles held in a liquid suspension.


The term “planar array” means an immunoassay system where individual biomarkers or binding agents are immobilised in discrete addressable locations on a continuous solid surface including but not limited to glass slide, silicon wafer, nitrocellulose strip. Subsequent steps of an immunoassay are then performed using common reagents applied to the entire surface of the planar array or may be added individually at the appropriate addressable location within the array.


The term “isolated”, or grammatical equivalents thereof, means throughout this specification, that the protein, antibody, polynucleotide or chemical molecule as the case may be, exists in a physical milieu distinct from that in which it may occur in nature.


As used herein, the term “subject” includes any human or non-human animal. The term “non-human animal” includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, rodents, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.


The term “treat”, “treating”, “treatment”, “prevent”, “preventing” or “prevention”, or grammatical equivalents thereof, includes therapeutic treatments, prophylactic treatments and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses the reduction of the symptoms or underlying risk factors.


The term “diagnosis”, or grammatical equivalents thereof, as used herein, includes the provision of any information concerning the existence or presence, non-existence or absence or probability of the disorder in a patient. It further includes the provision of information concerning the type or classification of the disorder or of symptoms which are or may be experienced in connection with it. This may include, for example, diagnosis of the severity of the disorder. It encompasses prognosis of the medical course of the disorder, for example its duration, severity and the course of progression from mild cognitive impairment (MCI) to AD or other dementias.


The term “staging”, or grammatical equivalents thereof, as used herein, means identifying in a subject the stage of a neurocognitive disorder, in particular AD. For example, AD is characterised by 3 stage or 7 stages, depending on the diagnostic framework used. The Global Dementia Scale is one such measure of global function. It is measured by assessment of severity including cognition and function against a standardised set of severity criteria.


The term “efficacy” indicates the capacity for beneficial change of a given intervention (e.g. a drug, medical device, surgical procedure, etc.). If efficacy is established, that intervention is likely to be at least as good as other available interventions, to which it will have been compared. The term “efficacy” and “effectiveness” are used herein interchangeably.


The term “comprising” indicates that the subject includes all the elements listed, but may, optionally, also include additional, unnamed elements (i.e. open).


The term “and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example “A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein.


Unless the context dictates otherwise, the definitions of the features/terms set out above are not limited to any particular aspect or embodiment of the invention and apply equally to all aspects and embodiments which are described herein.


Abbreviations

CSF (cerebrospinal fluid); LBD (Lewy body dementia); FTD (fronto-temporal dementia); VaD (vascular dementia); ALS (amyotrophic lateral sclerosis) CJD (Creutzfeldt-Jakob disease); CNS (central nervous system); TMT® (Tandem Mass Tag®); TEAB (Tetra-ethylamonium Bicarbonate); TFA (Trifluoroacetic acid); SDS (Sodium dodecyl sulfate); TCEP (Tris(2-carboxyethyl)phosphine); ACN (Acetonitrile); Da (Dalton); HPLC (High-performance liquid chromatography); FA (Formic acid); IFC (Intelligent flow control); LC-MS/MS (Liquid Chromatography with tandem Mass Spectrometry detection); MS (Mass Spectrometry); MS/MS or MS2 (Tandem MS); MS/MS/MS or MS3 (triple MS) PAGE (Polyacrylamide gel electrophoresis); SCX (Strong Cation Exchange); ppm (Parts per million); TiO2 (titanium dioxide); IMAC (iron metal affinity chromatography); ELISA (enzyme-linked immonusorbent assay).


DETAILED DESCRIPTION
1. Proteins Panels and Methods of Using Thereof

Whilst it is widely accepted that tau is involved in the pathology of neurodegenerative disorders such as tauopathies, like AD, and that it actively participates in the formation of neurofibrillary tangles, not much is known about the molecular processes that mediate toxicity of over expressed and hyperphosphorylated tau.


The present inventors hypothesized that tau over expression and hyperphosphorylation causes tau toxicity in the brain leading to further events, including activation of signalling pathways, which may contribute to the advancement and severity of the disease. They further hypothesised that many of the proteins involved in these pathways would be released into blood and CSF during the evolution of tau toxicity and would be detectable early in the disease process.


The present inventors have surprisingly found that multiple cellular processes, the associated proteins and/or their levels are modified in a tau dose-dependent manner and that these affected pathways are correlated with several hallmarks of AD.


Furthermore, the panel of biomarkers identified herein are not exclusively expressed in the brain, they are surprisingly also identifiable in the cerebrospinal fluid (CSF) and most importantly their abundance in CSF is regulated between non-AD and AD patients with substantive memory effects.


Finally, the present inventors successfully demonstrated herein that upon administration of tau kinase inhibitors (enzymes which inhibits tau phosphorylation), the abundance of the proteins (i.e. biomarkers) of the panels according to the present invention increases or decreases inversely to their pre-administration abundance, indicating that the inhibitors are effective in reducing hyperphosphorylation of tau. Most surprisingly, as the abundance of these proteins increases or decreases inversely to their pre-administration abundance, also the tau toxicity comparably decreases.


The present invention provides for a panel of biomarkers comprising:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • and/or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Protein phosphatase 1 regulatory subunit 14A is an inhibitor of PPP1CA (serine/threonine-protein phosphatase PP1-alpha catalytic subunit). Has over 1000-fold higher inhibitory activity when phosphorylated, creating a molecular switch for regulating the phosphorylation status of PPP1CA substrates and smooth muscle contraction.


Described to participate in RNA metabolism in the myelinating cell, 2′,3′-cyclic-nucleotide 3′-phosphodiesterase is the third most abundant protein in central nervous system myelin. Its catalytic activity is on nucleoside 2′,3′-cyclic phosphate which is transformed into nucleoside 2′-phosphate.


An isoform is described herein as an alternative protein sequence with respect to the canonical sequence. Isoforms can be generated from the same gene by a single or by the combination of alternative promoter usage, alternative splicing, alternative initiation and ribosomal frameshifting.


A variant is described herein as to include natural variants such as (naturally occurring) polymorphisms, variations between strains, isolates or cultivars, disease-associated mutations and RNA editing events. A variant is generally reported as the amino acid change with respect to the canonical sequence. Most naturally occurring polymorphisms (also called single amino acid polymorphisms or SAPs) are due to a single nucleotide change at the codon level. RNA editing events include conversion, insertion and deletion of nucleotides.


A fragment is described herein as the result of proteolytic (enzymatic or else) cleavage of a protein. Fragments may be the results of natural proteolytic cleavage for example fragments generated during the activation of complement, the clotting cascade, or from enzymatic cleavage of matrix proteins. Alternatively, fragments may be generated in-vivo and/or in-vitro for example with proteases.


In one embodiment, the variant of protein phosphatase 1 regulatory subunit 14A comprises or has the amino acid sequence of SEQ ID NO: 1 and wherein


a) serine at position 26 is replaced by phosphoserine; or


b) threonine at position 38 is replaced by phosphothreonine; or


c) serine at position 136 is replaced by phosphoserine; or


d) glycine at position 123 is replaced by arginine.



FIG. 1 shows the human sequence of protein phosphatase 1 regulatory subunit 14A (SEQ ID NO:1); flagged by symbol □ are those amino acids that are replaced with a different amino acid in isoforms of human protein phosphatase 1 regulatory subunit 14A as indicated above in d). Amino acids flagged by a * are those amino acids which are replaced by modified amino acids in isoforms of human protein phosphatase 1 regulatory subunit 14A as indicated above in a) to c).


In another embodiment the isoform of protein phosphatase 1 regulatory subunit 14A comprises or has the amino acid sequence of SEQ ID NO:1 wherein amino acid 68 to 94 are missing (isoform 2).


In another embodiment, the variant of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprises or has the amino acid sequence of SEQ ID NO: 2 and wherein


a) tyrosine at position 110 is replaced by phosphotyrosine; or


b) cysteine at position 418 is replaced by cysteine methyl ester; or


c) cysteine at position 418 is replaced by S-farnesyl cysteine; or


d) glutamine at position 207 is replaced by arginine.



FIG. 2 shows the human sequence of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase (SEQ ID NO:2); flagged by symbol □ are those amino acids that are replaced with a different amino acid in isoforms of human 2′,3′-cyclic-nucleotide 3′-phosphodiesterase as indicated above in d). Amino acids flagged by a * are those amino acids which are replaced by modified amino acids in isoforms of human protein phosphatase 1 regulatory subunit 14A as indicated above in a) to c).


In another embodiment the isoform of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprises or has the amino acid sequence of SEQ ID NO:2 wherein amino acid 1 to 20 are missing (isoform CNPI).


In general clinical practice, proteins for use as biomarkers are measured as a set of at least 2, preferably at least 3 or 4. Hence, in addition to protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof and/or 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof, the panel of biomarkers according to the present invention may further comprise one or more, alternative two or more, preferably more than two proteins selected from Group A which comprises Actin alpha cardiac muscle 1 comprising or having the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising or having the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising or having the amino acid sequence of SEQ ID NO:3, cAMP-dependent protein kinase type I-beta regulatory subunit comprising or having the amino acid sequence of SEQ ID NO:24, Catenin delta-1 comprising or having the amino acid sequence of SEQ ID NO:4, Centrosomal protein of 170 kDa comprising or having the amino acid sequence of SEQ ID NO:23, Clathrin light chain B comprising or having the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising or having the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising or having the amino acid sequence of SEQ ID NO:14, GMP reductase 1 comprising or having the amino acid sequence of SEQ ID NO:27, Guanine nucleotide-binding protein G(q) subunit alpha comprising or having the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising or having the amino acid sequence of SEQ ID NO:15, KxDL motif-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:28, Lambda-crystallin homolog comprising or having the amino acid sequence of SEQ ID NO:18, Myelin-associated oligodendrocyte basic protein comprising or having the amino acid sequence of SEQ ID NO:20, Neutral alpha-glucosidase AB comprising or having the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising or having the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:16, Protein KIAA1045 comprising or having the amino acid sequence of SEQ ID NO:25, Secernin-2 comprising or having the amino acid sequence of SEQ ID NO:8, Serum albumin comprising or having the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising or having the amino acid sequence of SEQ ID NO:9, Synaptoporin comprising or having the amino acid sequence of SEQ ID NO:22, Syntaphilin comprising or having the amino acid sequence of SEQ ID NO:10, Transmembrane protein 119 comprising or having the amino acid sequence of SEQ ID NO: 21 and Tubulin alpha chain-like 3 comprising or having the amino acid sequence of SEQ ID NO:26.


Table 1 shows the names of the biomarkers of “Group A” and their Uniprot codes for the human protein and its mouse counterpart.









TABLE 1







Biomarkers of Group A










SEQ

UNIPROT
UNIPROT


ID

ID
ID


NO:
Protein Name
(human)
(mouse)













1
phosphatase 1 regulatory subunit 14A
Q96A00
Q91VC7


2
2′,3′-cyclic-nucleotide 3′-
P09543
P16330



phosphodiesterase


3
BH3-interacting domain death agonist
P55957
P70444


4
Catenin delta-1
O60716
P30999


5
Clathrin light chain B
P09497
Q61RU5


6
Guanine nucleotide-binding protein G(q)
P50148
P21279



subunit alpha


7
Neutral alpha-glucosidase AB
Q14697
Q8BHN3


8
Secernin-2
Q96FV2
Q8VCA8


9
Short-chain specific acyl-CoA
P16219
Q07417



dehydrogenase, mitochondrial


10
Syntaphilin
O15079
Q80U23


11
Actin, alpha cardiac muscle 1
P68032
P68033


12
Antithrombin-III
P01008
P32261


13
Egl nine homolog 1
Q9GZT9
Q91YE3


14
Fibrinogen gamma chain
P02679
Q8VCM7


15
Insulin-like growth factor-binding protein
P24592
P47880


16
OCIA domain-containing protein 1
Q9NX40
Q9CRD0


17
Serum albumin
P02768
P07724


18
Lambda-crystallin homolog
Q9Y2S2
Q99KP3


19
Nuclear pore complex protein Nup155
O75694
Q99P88


20
Myelin-associated oligodendrocyte basic
Q13875
Q9D2P8



protein


21
Transmembrane protein 119
Q4V9L6
Q8R138


22
Synaptoporin
Q8TBG9
Q8BGN8


23
Centrosomal protein of 170 kDa
Q5SW79
H7BX26


24
cAMP-dependent protein kinase type 1
P31321
P12849


25
Protein KIAA1045
Q9UPV7
Q80TL4


26
Tubulin alpha chain-like 3
A6NHL2
Q3UX10


27
GMP reductase 1
P36959
Q9DCZ1


28
KxDL motif-containing protein 1
Q9BQD3
Q80XH1









In one embodiment, the panel of biomarkers according to the present invention comprises i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof and one or more, preferably two or more, biomarkers selected from “Group B”, which comprises Actin alpha cardiac muscle 1 comprising or having the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising or having the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising or having the amino acid sequence of SEQ ID NO:3, Catenin delta-1 comprising or having the amino acid sequence of SEQ ID NO:4, Clathrin light chain B comprising or having the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising or having the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising or having the amino acid sequence of SEQ ID NO:14, Guanine nucleotide-binding protein G(q) subunit alpha comprising or having the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising or having the amino acid sequence of SEQ ID NO:15, Lambda-crystallin homolog comprising or having the amino acid sequence of SEQ ID NO:18, Neutral alpha-glucosidase AB comprising or having the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising or having the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:16, Secernin-2 comprising or having the amino acid sequence of SEQ ID NO:8, Serum albumin comprising or having the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising or having the amino acid sequence of SEQ ID NO:9 and Syntaphilin comprising or having the amino acid sequence of SEQ ID NO:10.


This sub-group of biomarkers (referred herein as “Group B”) have been identified in separate in human plasma or human serum of non-AD individuals (http://www.plasmaproteomedatabase.org/). Hence, it is to be expected that these biomarkers will be not only present in blood/blood products in AD patients but also that their up/down-regulation observed in CSF will translate in the blood/blood products. This is particularly advantageous as blood and its products (plasma or serum) are more readily and easily accessible for diagnosis than CSF.


In another embodiment, the panel of biomarkers comprises i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof and one or more, preferably two or more, biomarkers selected from “Group C”, which comprises Actin alpha cardiac muscle 1 comprising or having the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising or having the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising or having the amino acid sequence of SEQ ID NO:3, cAMP-dependent protein kinase type I-beta regulatory subunit comprising or having the amino acid sequence of SEQ ID NO:24, Catenin delta-1 comprising or having the amino acid sequence of SEQ ID NO:4, Centrosomal protein of 170 kDa comprising or having the amino acid sequence of SEQ ID NO:23, Clathrin light chain B comprising or having the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising or having the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising or having the amino acid sequence of SEQ ID NO:14, GMP reductase 1 comprising or having the amino acid sequence of SEQ ID NO:27, Guanine nucleotide-binding protein G(q) subunit alpha comprising or having the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising or having the amino acid sequence of SEQ ID NO:15, Lambda-crystallin homolog comprising or having the amino acid sequence of SEQ ID NO:18, Myelin-associated oligodendrocyte basic protein comprising or having the amino acid sequence of SEQ ID NO:20, Neutral alpha-glucosidase AB comprising or having the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising or having the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:16, Protein KIAA1045 comprising or having the amino acid sequence of SEQ ID NO:25, Serum albumin comprising or having the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising or having the amino acid sequence of SEQ ID NO:9, Synaptoporin comprising or having the amino acid sequence of SEQ ID NO:22, Syntaphilin comprising or having the amino acid sequence of SEQ ID NO:10 and Tubulin alpha chain-like 3 comprising or having the amino acid sequence of SEQ ID NO:26.


This sub-group of biomarkers (which will be referred herein as “Group C”) has been found to be regulated in human CSF of AD patients versus control subjects and inversely regulated in mouse brain upon administration of a casein kinase inhibitors.









TABLE 2







Biomarkers of “Group C”.












Human CSF
Mouse brain




AD/cntrl
Log 2


SEQ

Log 2
Ck1


ID
UNIPROT
Braak stage
inhibitor













NO:
ID
Protein Name
3/4
5/6
PS110
PS278
















1
Q96A00
phosphatase 1 regulatory
0.79
0.69
0.71
0.49




subunit 14A


2
P09543
2′,3′-cyclic-nucleotide 3′-
0.57
0.57
0.63
0.62




phosphodiesterase


3
P55957
BH3-interacting domain death
0.80

−0.53
−0.56




agonist


4
O60716
Catenin delta-1

0.58
−2.26
−1.07


5
P09497
Clathrin light chain B
0.90

0.70
0.50


6
P50148
Guanine nucleotide-binding
0.59

0.82
0.62




protein G(q) subunit alpha


7
Q14697
Neutral alpha-glucosidase AB
0.72
0.59
0.52
−0.78


9
P16219
Short-chain specific acyl-
−0.75

0.54
0.57




CoA dehydrogenase,




mitochondrial


10
O15079
Syntaphilin

0.52
−0.71
−0.76


11
P68032
Actin, alpha cardiac muscle 1
0.56

0.58



12
P01008
Antithrombin-III
−0.59

0.57



13
Q9GZT9
Egl nine homolog 1
0.77


−073


14
P02679
Fibrinogen gamma chain
−0.79

1.01



15
P24592
Insulin-like growth factor-
0.86

−0.53





binding protein


16
Q9NX40
OCIA domain-containing
0.77
0.94

−0.59




protein 1


17
P02768
Serum albumin
−0.63

0.69



18
Q9Y2S2
Lambda-crystallin homolog
0.60


0.56


19
O75694
Nuclear pore complex protein
0.49

−0.53





Nup155


20
Q13875
Myelin-associated
0.60

0.84
0.66




oligodendrocyte basic protein


22
Q8TBG9
Synaptoporin
0.91
1.03

0.72


23
Q5SW79
Centrosomal protein of 170 kDa
0.91
0.92

−0.51


24
P31321
cAMP-dependent protein kinase
1.56

0.53





type 1


25
Q9UPV7
Protein KIAA1045
0.67

−0.63



26
A6NHL2
Tubulin alpha chain-like 3
1.75


0.67


27
P36959
GMP reductase 1
1.30

0.68










In another embodiment the panel of biomarkers according to the present invention comprises i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof and one or more, preferably two or more, biomarkers selected from “Group D”, which comprises Secernin-2 comprising or having the amino acid sequence of SEQ ID NO:8, Transmembrane protein 119 comprising or having the amino acid sequence of SEQ ID NO: 21 KxDL motif-containing protein 1 comprising or having the amino acid sequence of SEQ ID NO:28.


This sub-group of biomarkers (which will be referred herein as “Group D”) has been found to be regulated in the brain of AD patients versus control and regulated in mouse brain upon administration of casein kinase inhibitors.









TABLE 3







Biomarkers of “Group D”.














Human
Mouse brain


SEQ


brain
Log 2


ID
UNIPROT

AD/cntrl
Ck1 inhibitor












NO:
ID
Protein Name
Log 2
PS110
PS278















1
Q96A00
phosphatase 1 regu-
0.79
0.69
0.71




latory subunit 14A


2
P09543
2′,3′-cyclic-nucleotide
0.57
0.57
0.63




3′-phosphodiesterase


8
Q96FV2
Secernin-2
0.56
−0.76
−0.52


21
Q4V9L6
Transmembrane
1.15
−0.66
−0.52




protein 119


28
Q9BQD3
KxDL motif-con-
0.58
−0.58





taining protein 1









Groups C and D (listed in Tables 2 and 3, respectively) comprise biomarkers which have been shown to be regulated in mouse brain following administration of two different casein kinase inhibitors and i) (Table 2; Group C) biomarkers which are up or down-regulated in the CSF of human patient with AD at Braak stages 3/4 or 5/6 or ii) (Table 3; Group D) biomarkers which are up/down-regulated in human brain of AD patients versus control subjects. These biomarkers are very advantageous as they allow to diagnose and stage the tauopathy but also serve as pharmacodynamics biomarkers allowing to establish if a treatment is effective in that specific patient or if an alternative approach should be followed.


Furthermore, the present invention provides for a panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422;


      wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


The microtubule-associated protein tau becomes abnormally phosphorylated in the hippocampus and cortex of patients with Alzheimer's disease, ultimately forming aggregates which organise into paired helical filaments (PHF tau), a pathological hallmark of the condition. Certain phosphorylated forms of tau protein can propagate through the brain by trans-synaptic spreading and presumably through this and other poorly understood processes is also found in cerebrospinal fluid. The ratio of tau phosphorylated at serine 181 (based on 2N4R tau isoform sequence; SEQ ID NO: 29) relative to total tau within the CSF is an accepted biomarker used to classify individuals as having pre-symptomatic Alzheimer's disease and to confirm the clinical diagnosis in symptomatic disease.


The amino acids phosphorylated on tau having or comprising the amino acid sequence of SEQ ID NO: 29 or fragments thereof identified in the present inventions are serine and/or threonine and/or tyrosine amino acids as illustrated in Table 4.









TABLE 4







Amino acids of tau protein (SEQ ID NO: 29) found


to be phosphorylated in tau from human CSF and/or


human brain and/or mouse brain (X means detected)










Amino acid Number
Mouse Brain
Human Brain
Human CSF





T39
X




S46
X
X
X


T50
X
X


T52
X


T56
X
X


S61
X
X
X


T63
X


S64
X

X


S68
X


T69
X


S113
X


T181
X
X
X


S184
X
X
X


S185
X
X
X


S191
X
X


S195
X


S198
X
X
X


S199
X
X
X


S202
X
X
X


S205
X
X
X


S208
X
X
X


S210
X
X


T212
X
X
X


S214
X
X
X


T217
X
X
X


T231
X
X
X


S235

X
X


S237

X
X


S238

X
X


S258

X
X


S262
X
X
X


S285


X


S289

X
X


S356
X
X
X


Y394
X
X


S396
X
X
X


S400
X
X
X


T403
X
X
X


S404

X
X


S409
X
X


S412
X


S413
X


T414/S416
X


S422

X









Each of the amino acid shown in Table 4 have diagnostic, prognostic and/or pharmacodynamic biomarker utility. Preferably the seventeen phosphorylated residues that were detected in all studies (X in all columns) have greatest potential as pharmacodynamics biomarkers as they are present during human disease, are also detected in an animal model treated with a casein kinase inhibitor, and could also be detected in CSF of humans.


More preferably, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64, T181, S184, S202, S205, T231 and/or S235.


In one embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from S61, S64, S199, S205, and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acid S61.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acid S64.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acid S199.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acid S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acid S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61 and S64.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61 and S199.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64 and S199.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S199 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S199 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S205 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64 and S199.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S199 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S199 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S205 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64, S199 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64, S199 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S64, S205 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S199, S205 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64, S199 and S205.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64, S199 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S199, S205 and S396.


In another embodiment, the panel of biomarkers comprising tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises phosphorylated amino acids S61, S64, S199, S205, and S396.


The embodiments relating to the amino acid sequence and the particular phosphorylated amino acids in tau or on one or more fragments thereof, as described above, are equally applicable to all other embodiments of the second aspect of the invention and to all other aspects of the invention where tau or on one or more fragments thereof is involved.


In one embodiment, the panel of biomarker comprises at least one biomarker selected from Groups A, B, C or D and one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422;


      wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid. Preferably, the biomarker selected from Groups A, B, C or D is:
    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


More preferably, the panel comprises three or more, four or more, five or more biomarkers, wherein at least two biomarkers, in addition to tau, are selected from Groups A, B, C or D and are:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • and
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Additionally, the present invention provides for a panel of biomarkers comprising one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof.


These biomarkers have been found to be highly regulated in the CSF of AD patients versus control. All non-modified peptides for each protein were summed for three control and three AD cases. The log 2 ratio and p-value of each protein was then calculated. Proteins with 2 or more peptides, >60% regulation and p<0.05 were selected as biomarkers for Table 5.









TABLE 5







Biomarkers up/down-regulated in the


CSF of AD patient versus control












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













I3L192
Basigin
−3.86
0.04


E5RJZ1
Cytochrome c oxidase subunit 7A-
4.19
0.05



related protein, mitochondrial


K7EIT4
14-3-3 protein epsilon
0.78
0.04


K7EJ68
3-ketoacyl-CoA thiolase,
0.91
0.01



mitochondrial


B4DVF4
3-ketoacyl-CoA thiolase, peroxisomal
1.28
0.02


P25325
3-mercaptopyruvate sulfurtransferase
0.81
0.01


P08708
40S ribosomal protein S17
1.34
0.01


E5RIP1
40S ribosomal protein S20
1.32
0.01


D6R9I7
40S ribosomal protein S23
0.83
0.04


P61247
40S ribosomal protein S3a
0.96
0.04


F5GZI0
4F2 cell-surface antigen heavy chain
1.29
0.03


P32754
4-hydroxyphenylpyruvate dioxygenase
0.76
0.03


F8VPE8
60S acidic ribosomal protein P0
0.73
0.03


H7C3M2
60S ribosomal protein L3
2.45
0.01


C9JIJ5
60S ribosomal protein L7
0.86
<0.01


Q8IZP0
Abl interactor 1
1.04
0.01


H0YN26
Acidic leucine-rich nuclear
1.76
0.04



phosphoprotein 32 family member A


E9PF58
Actin-related protein 2/3 complex
0.94
0.02



subunit 1A


B4DXW1
Actin-related protein 3
0.73
0.04


I3L1U8
Active breakpoint cluster region-
0.92
<0.01



related protein


O14561
Acyl carrier protein, mitochondrial
0.72
0.02


P07108
Acyl-CoA-binding protein
1.18
0.02


O14734
Acyl-coenzyme A thioesterase 8
0.76
<0.01


E9PQQ8
Adenylate kinase isoenzyme 5
0.89
0.02


P12235
ADP/ATP translocase 1
0.71
0.03


F5H1V1
ADP-ribosylation factor 3
0.74
0.03


Q9NVJ2
ADP-ribosylation factor-like protein
0.94
0.01



8B


H0Y5U1
Agrin
1.05
0.03


P11766
Alcohol dehydrogenase class-3
0.87
0.05


A6NHU4
Aldo-keto reductase family 1 member
−1.04
0.01



C1


P55008
Allograft inflammatory factor 1
0.82
0.03


P02763
Alpha-1-acid glycoprotein 1
−1.03
0.02


G3V3A0
Alpha-1-antichymotrypsin
−0.84
0.04


H0YJ11
Alpha-actinin-1
0.67
0.01


M0R2M1
Alpha-soluble NSF attachment protein
0.74
0.03


P05067
Amyloid beta A4 protein
0.90
<0.01


K7EMN4
Amyloid-like protein 1
0.73
0.01


E9PQS3
Amyloid-like protein 2
0.94
0.01


P03950
Angiogenin
−0.76
0.03


E7EV01
Ankyrin repeat and SOCS box protein 2
1.68
0.01


P16157
Ankyrin-1
0.86
0.01


D6R9U4
Ankyrin-2
0.65
0.03


V9GYC1
Apolipoprotein A-II
−0.85
<0.01


P06727
Apolipoprotein A-IV
−1.97
0.04


P04114
Apolipoprotein B-100
0.70
0.04


B0YIW2
Apolipoprotein C-III
−0.77
0.04


G3V1B6
Apolipoprotein O
0.84
0.03


H0YER7
Apoptosis inhibitor 5
0.79
0.04


E9PMA0
Apoptosis-inducing factor 1,
0.69
0.03



mitochondrial


P29972
Aquaporin-1
0.90
<0.01


B0YIW6
Archain 1
−1.26
0.01


F8VXI9
ARF GTPase-activating protein GIT2
1.10
0.02


Q96P47
Arf-GAP with GTPase, ANK repeat and
0.66
0.01



PH domain-containing protein 3


O43776
Asparagine--tRNA ligase, cytoplasmic
1.37
0.03


P17174
Aspartate aminotransferase,
0.99
0.03



cytoplasmic


B1AJS1
Astrotactin-1
0.70
0.01


K7EJP1
ATP synthase subunit alpha,
0.77
0.04



mitochondrial


C9JU26
ATP synthase subunit f, mitochondrial
0.75
0.03


P17858
ATP-dependent 6-phosphofructokinase,
0.68
0.02



liver type


Q96BJ3
Axin interactor, dorsalization-
0.98
0.04



associated protein


Q4VXN1
Band 4.1-like protein 1
1.36
0.01


E9PQD2
Band 4.1-like protein 2
3.48
0.02


P02749
Beta-2-glycoprotein 1
−0.70
0.00


Q562R1
Beta-actin-like protein 2
0.77
0.05


P35612
Beta-adducin
0.77
0.05


Q96KN2
Beta-Ala-His dipeptidase
0.71
0.05


P07686
Beta-hexosaminidase subunit beta
1.49
0.00


C9JSN9
Biotinidase
0.97
0.01


Q9NZE6
Eukaryotic translation initiation
0.75
0.01



factor 4A, isoform 2, isoform CRA_b


Q8WXS3
Brain and acute leukemia cytoplasmic
0.88
0.05



protein


P11274
Breakpoint cluster region protein
0.99
0.01


Q96GW7
Brevican core protein
0.69
0.03


Q9HCU4
Cadherin EGF LAG seven-pass G-type
0.68
<0.01



receptor 2


P19022
Cadherin-2
1.17
0.05


Q9UQM7
Calcium/calmodulin-dependent protein
0.81
0.02



kinase type II subunit alpha


E7ETC9
Calcium/calmodulin-dependent protein
1.08
<0.01



kinase type II subunit beta


Q16566
Calcium/calmodulin-dependent protein
0.85
0.03



kinase type IV


H7C4P2
Calcium-dependent secretion activator
0.87
0.03



1


F8WBR5
Calmodulin
1.09
0.03


Q9P1Y5
Calmodulin-regulated spectrin-
0.91
0.01



associated protein 3


P07384
Calpain-1 catalytic subunit
1.06
0.03


O94985
Calsyntenin-1
0.75
0.01


Q9BQT9
Calsyntenin-3
0.76
0.01


O43852
Calumenin
1.15
0.01


C9J9E2
CaM kinase-like vesicle-associated
0.70
0.05



protein


K7EM13
cAMP-dependent protein kinase type I-
0.97
<0.01



alpha regulatory subunit


P31321
cAMP-dependent protein kinase type I-
1.56
0.04



beta regulatory subunit


Q5TDF0
Cancer-related nucleoside-
0.93
0.01



triphosphatase


Q8WXD9
Caskin-1
0.76
0.04


Q92851
Caspase-10
1.21
0.01


Q86VU5
Catechol O-methyltransferase domain-
1.06
0.01



containing protein 1


C9IZ88
Catenin alpha-2
2.11
0.01


E9PKT6
Cathepsin H
0.73
0.01


E9PNW4
CD59 glycoprotein
0.79
0.04


Q5JYX0
Cell division control protein 42
1.04
0.05



homolog


Q99674
Cell growth regulator with EF hand
0.86
<0.01



domain protein 1


Q5SW79
Centrosomal protein of 170 kDa
0.92
0.02


Q8NI60
Chaperone activity of bc1 complex-
0.78
0.01



like, mitochondrial


Q7LBR1
Charged multivesicular body protein
0.66
0.01



1b


Q96FZ7
Charged multivesicular body protein 6
0.83
<0.01


Q9BWS9
Chitinase domain-containing protein 1
0.85
0.02


J3KS05
Chromobox protein homolog 1
0.84
0.01


G5E968
Chromogranin A, isoform CRA_b
0.78
0.01


Q9UPT6
C-Jun-amino-terminal kinase-
−0.85
0.01



interacting protein 3


Q8IZR5
CKLF-like MARVEL transmembrane
0.70
0.01



domain-containing protein 4


E5RGY9
Clathrin coat assembly protein AP180
1.45
0.02


D6RJD1
Clathrin light chain B
0.84
0.02


Q14019
Coactosin-like protein
0.68
0.02


P00740
Coagulation factor IX
−0.73
0.05


P53618
Coatomer subunit beta
1.07
0.03


H0Y8X7
Coatomer subunit gamma-1
0.69
0.03


I3L0M4
Coiled-coil domain-containing protein
0.99
0.01



43


Q96A33
Coiled-coil domain-containing protein
0.79
0.02



47


H7C5H1
Complement factor B
−1.11
0.03


Q03591
Complement factor H-related protein 1
−0.67
<0.01


V9GYE7
Complement factor H-related protein 2
−1.01
0.01


E9PDN6
Contactin-associated protein-like 4
0.85
0.01


H0YKU5
COP9 signalosome complex subunit 2
1.31
0.01


Q92905
COP9 signalosome complex subunit 5
0.76
0.01


O75367
Core histone macro-H2A.1
0.67
0.01


H0YJG0
Creatine kinase B-type
0.68
0.03


E7EPF8
C-terminal-binding protein 1
0.84
0.01


E9PC62
CUGBP Elav-like family member 2
0.99
0.01


F5H6I6
Cullin-associated NEDD8-dissociated
1.06
0.04



protein 1


E9PHZ2
Cysteine and histidine-rich domain-
0.67
0.02



containing protein 1


Q16878
Cysteine dioxygenase type 1
1.32
0.05


H0YFA4
Cysteine-rich protein 2
0.95
0.03


H3BP04
Cytochrome b-c1 complex subunit 2,
0.67
0.02



mitochondrial


P14927
Cytochrome b-c1 complex subunit 7
0.98
0.05


P47985
Cytochrome b-c1 complex subunit
1.13
0.04



Rieske, mitochondrial


Q5JTJ3
Cytochrome c oxidase assembly factor
1.23
0.01



6 homolog


K7EQD3
Cytochrome c oxidase subunit 6B1
0.88
0.02


Q99418
Cytohesin-2
0.77
0.03


E7EPF5
Cytoplasmic protein NCK2
0.72
0.02


C9JM78
D-beta-hydroxybutyrate dehydrogenase,
1.46
0.00



mitochondrial


Q9UKG1
DCC-interacting protein 13-alpha
0.79
0.01


H7C342
D-dopachrome decarboxylase
0.88
0.03


Q8NFT8
Delta and Notch-like epidermal growth
1.02
0.01



factor-related receptor


Q08495
Dematin
0.76
0.01


P60981
Destrin
0.93
0.05


Q9BPU6
Dihydropyrimidinase-related protein 5
0.82
0.03


E7EPF1
Disintegrin and metalloproteinase
1.05
0.01



domain-containing protein 22


B4E2H8
Disks large homolog 1
1.37
0.03


A8MVA8
Disks large homolog 2
0.72
0.02


O00273
DNA fragmentation factor subunit
−0.67
0.03



alpha


O75190
DnaJ homolog subfamily B member 6
1.27
<0.01


Q9NVH1
DnaJ homolog subfamily C member 11
0.94
0.01


Q13217
DnaJ homolog subfamily C member 3
0.86
<0.01


K7EIH8
DnaJ homolog subfamily C member 7
0.92
0.02


B7Z4L4
Dolichyl-diphosphooligosaccharide--
1.87
0.01



protein glycosyltransferase subunit 1


Q9P0K9
DOMON domain-containing protein
1.48
0.01



FRRS1L


F8VYL3
Dynamin-1-like protein
0.89
0.01


P50570
Dynamin-2
0.89
0.03


Q9P225
Dynein heavy chain 2, axonemal
1.15
0.00


H3BS86
E3 ubiquitin-protein ligase CHIP
0.83
0.01


Q05639
Elongation factor 1-alpha 2
0.68
0.04


H3BNU3
Elongation factor Tu, mitochondrial
1.14
0.01


Q9NZ08
Endoplasmic reticulum aminopeptidase
1.16
<0.01



1


H0YIV0
Endoplasmin
1.18
0.04


B1AKC9
Ephrin type-B receptor 2
1.26
0.02


P20827
Ephrin-A1
0.86
0.05


Q92506
Estradiol 17-beta-dehydrogenase 8
0.87
0.04


Q92731
Estrogen receptor beta
1.73
0.01


E7EMV8
Eukaryotic initiation factor 4A-II
−0.98
0.03


A6NJH9
Eukaryotic translation initiation
0.91
0.03



factor 1A, Y-chromosomal


O75821
Eukaryotic translation initiation
1.02
0.04



factor 3 subunit G


O43909
Exostosin-like 3
1.03
<0.01


I3L252
FAD-AMP lyase
3.43
0.02


H3BRW3
FAD-linked sulfhydryl oxidase ALR
1.50
0.01


C9JPH9
Fascin
0.86
0.01


G3V1D1
Ferritin
1.37
0.03


Q6MZW2
Follistatin-related protein 4
1.38
<0.01


H3BR68
Fructose-bisphosphate aldolase A
0.93
<0.01


H6UMI1
Gamma-aminobutyric acid receptor-
1.40
0.01



associated protein


H0YJU6
Gamma-aminobutyric acid receptor
0.78
0.01



subunit beta-3


E5RGR6
GDNF family receptor alpha-2
1.06
<0.01


G3V582
Gephyrin
1.07
<0.01


K7EMP8
Glial fibrillary acidic protein
0.66
0.03


Q8TDQ7
Glucosamine-6-phosphate isomerase 2
−0.65
0.04


K7EJ70
Glucosidase 2 subunit beta
0.80
<0.01


P42261
Glutamate receptor 1
1.13
0.04


G3V164
Glutamate receptor 4
1.02
0.01


F5H4N6
Glutamate receptor-interacting
−0.87
<0.01



protein 1


H7BZD1
Glutaminase kidney isoform,
0.79
0.03



mitochondrial


O76003
Glutaredoxin-3
0.90
0.04


P07203
Glutathione peroxidase 1
1.13
0.02


Q03013
Glutathione S-transferase Mu 4
1.09
0.04


P78417
Glutathione S-transferase omega-1
1.13
0.01


H0YMX4
Glycine amidinotransferase,
1.67
0.04



mitochondrial


P23434
Glycine cleavage system H protein,
0.94
0.01



mitochondrial


H7C024
Glypican-1
1.05
<0.01


Q9NZH0
G-protein coupled receptor family C
0.97
0.03



group 5 member B


P01112
GTPase HRas
1.09
0.01


Q9Y2T3
Guanine deaminase
0.75
0.01


B1AM21
Guanine nucleotide-binding protein
0.76
0.04



G(q) subunit alpha


P63092
Guanine nucleotide-binding protein
1.00
0.03



G(s) subunit alpha isoforms short


B7Z685
Guanylate cyclase 1, soluble, beta 3,
−0.64
0.02



isoform CRA_c


B1ANH6
Guanylate kinase
0.69
0.01


P00738
Haptoglobin
−1.07
<0.01


P00739
Haptoglobin-related protein
−1.65
<0.01


Q6PIK3
HCG1995540, isoform CRA_b
0.65
0.03


D6RG00
HCG2018358, isoform CRA_d
0.75
<0.01


H3BQZ7
HCG2044799
0.69
0.01


K7ENF6
Heat shock 70 kDa protein 12A
0.86
0.04


P08107
Heat shock 70 kDa protein 1A/1B
0.70
0.03


R4GN69
Heat shock protein 105 kDa
−0.83
0.01


C9J3N8
Heat shock protein beta-1
−0.91
0.01


Q5H962
HECT, UBA and WWE domain containing 1
1.00
0.05


Q15477
Helicase SKI2W
0.86
0.04


F5GWX2
Heme-binding protein 1
1.24
<0.01


P02100
Hemoglobin subunit epsilon
1.50
0.01


Q6ZVN8
Hemojuvelin
0.87
<0.01


Q8IZP7
Heparan-sulfate 6-O-sulfotransferase
0.78
0.01



3


P05546
Heparin cofactor 2
−0.66
0.03


Q9Y3E1
Hepatoma-derived growth factor-
−0.67
0.04



related protein 3


Q13151
Heterogeneous nuclear
0.90
0.03



ribonucleoprotein A0


H0YB39
Heterogeneous nuclear
0.80
0.02



ribonucleoprotein H


M0QYQ7
Heterogeneous nuclear
0.67
0.02



ribonucleoprotein M


B1AR61
Hexokinase-1
0.83
0.02


D6RD60
Histidine triad nucleotide-binding
1.01
0.03



protein 1


P16403
Histone H1.2
1.33
0.05


D6RCF2
Histone H2A
0.83
0.01


U3KQK0
Histone H2B
0.73
0.03


B4DEB1
Histone H3
1.31
0.03


Q6NXT2
Histone H3.3C
1.27
0.04


Q16775
Hydroxyacylglutathione hydrolase,
0.85
0.04



mitochondrial


Q9Y4L1
Hypoxia up-regulated protein 1
0.86
0.02


P22304
Iduronate 2-sulfatase
1.30
0.03


A6NGN9
IgLON family member 5
1.24
<0.01


H0Y4R1
Inosine-5′-monophosphate
0.83
0.04



dehydrogenase 2


H0YB38
Inositol monophosphatase 3
1.12
0.04


Q9UMF0
Intercellular adhesion molecule 5
0.80
0.01


K7EKJ9
Interleukin enhancer-binding factor 3
1.57
0.04


C9J826
Junction plakoglobin
2.03
0.04


P13645
Keratin, type I cytoskeletal 10
0.66
0.02


O43790
Keratin, type II cuticular Hb6
0.66
0.02


E9PES4
Kinesin-like protein KIF3A
0.67
0.05


P01042
Kininogen-1
−0.67
0.02


Q04760
Lactoylglutathione lyase
0.71
0.04


E9PLW6
L-aminoadipate-semialdehyde
−1.29
<0.01



dehydrogenase-phosphopantetheinyl



transferase


E5RH50
La-related protein 1
0.66
<0.01


F6S6P2
Large proline-rich protein BAG6
0.89
0.03


Q15334
Lethal(2) giant larvae protein
0.74
0.04



homolog 1


Q9UIC8
Leucine carboxyl methyltransferase 1
1.10
<0.01


H0YMX3
Leucine-rich repeat and
1.28
<0.01



immunoglobulin-like domain-containing



nogo receptor-interacting protein 1


Q6F5E8
Leucine-rich repeat-containing
−1.05
0.02



protein 16C


M0R2G0
Leucine-rich repeat-containing
0.76
<0.01



protein 4B


G3V1D4
Lin-7 homolog C (C. elegans), isoform
0.72
0.05



CRA_b


Q5VVL7
Lipoamide acyltransferase component
0.84
0.03



of branched-chain alpha-keto acid



dehydrogenase complex, mitochondrial


C9JXK9
Lipoma-preferred partner
1.08
0.01


E9PJZ7
Liprin-alpha-1
1.21
0.03


Q13136
Liprin-alpha-1
0.85
0.01


G3V200
Liprin-alpha-2
1.52
0.04


A8MW50
L-lactate dehydrogenase
0.72
0.02


F5GZQ4
L-lactate dehydrogenase A chain
0.68
<0.01


O95573
Long-chain-fatty-acid--CoA ligase 3
0.82
0.01


P10619
Lysosomal protective protein
0.70
0.02


F8VV32
Lysozyme C
−0.99
0.02


Q9NZW5
MAGUK p55 subfamily member 6
0.72
0.02


A2A2V1
Major prion protein
1.20
<0.01


C9JF79
Malate dehydrogenase
1.07
0.04


F5GX14
Malectin
0.75
0.02


P49006
MARCKS-related protein
0.95
0.03


B3KM87
Matrin-3
0.70
0.02


Q8N3F0
Maturin
0.89
0.02


Q5HYI7
Metaxin-3
1.07
<0.01


F8VSC4
Methionine aminopeptidase 2
0.65
0.01


Q13825
Methylglutaconyl-CoA hydratase,
1.50
0.03



mitochondrial


D6RCL2
Microtubule-associated protein 1B
0.73
0.04


M0QXQ9
Microtubule-associated protein 1S
0.76
0.02


Q9H936
Mitochondrial glutamate carrier 1
0.66
<0.01


G3V502
Mitochondrial import inner membrane
0.92
0.03



translocase subunit Tim9


E5RJK1
Mitochondrial peptide methionine
0.87
<0.01



sulfoxide reductase


Q10713
Mitochondrial-processing peptidase
0.70
0.04



subunit alpha


D6RAU3
Mitogen-activated protein kinase 10
0.65
0.04


Q15746
Myosin light chain kinase, smooth
1.08
0.01



muscle


E7ERA5
Myosin-10
0.77
0.02


P58546
Myotrophin
0.79
0.04


O95865
N(G),N(G)-dimethylarginine
0.83
0.01



dimethylaminohydrolase 2


O14745
Na(+)/H(+) exchange regulatory
1.06
0.05



cofactor NHE-RF1


Q4G0N4
NAD kinase 2, mitochondrial
0.96
<0.01


D6RAI5
NAD(P) transhydrogenase,
0.86
<0.01



mitochondrial


F8VRD8
NADH dehydrogenase [ubiquinone] 1
1.00
0.05



alpha subcomplex subunit 12


O95182
NADH dehydrogenase [ubiquinone] 1
0.73
0.02



alpha subcomplex subunit 7


Q16795
NADH dehydrogenase [ubiquinone] 1
0.74
0.04



alpha subcomplex subunit 9,



mitochondrial


E9PQ68
NADH dehydrogenase [ubiquinone] 1
−0.85
<0.01



beta subcomplex subunit 8,



mitochondrial


E7EPT4
NADH dehydrogenase [ubiquinone]
0.92
0.05



flavoprotein 2, mitochondrial


P56181
NADH dehydrogenase [ubiquinone]
0.70
0.03



flavoprotein 3, mitochondrial


Q9UHQ9
NADH-cytochrome b5 reductase 1
0.72
0.02


Q9BXJ9
N-alpha-acetyltransferase 15, NatA
0.95
0.04



auxiliary subunit


Q8TBC4
NEDD8-activating enzyme E1 catalytic
0.82
0.01



subunit


Q59FP8
Neogenin
0.98
0.04


O00533
Neural cell adhesion molecule L1-like
0.80
<0.01



protein


E7EQN4
Neurexin-1-beta
0.99
<0.01


H7C2R8
Neurexin-2
1.11
<0.01


Q9Y4C0
Neurexin-3
0.93
<0.01


Q9HDB5
Neurexin-3-beta
0.98
<0.01


Q9NPD7
Neuritin
0.94
0.03


P61601
Neurocalcin-delta
1.33
0.03


Q9UBB6
Neurochondrin
0.80
0.02


P16519
Neuroendocrine convertase 2
1.52
<0.01


Q8NFZ4
Neuroligin-2
0.96
0.04


O95502
Neuronal pentraxin receptor
1.09
<0.01


P47972
Neuronal pentraxin-2
1.02
0.01


O15240
Neurosecretory protein VGF
1.12
<0.01


C9JQU8
Neuroserpin
0.71
0.03


F5H810
Noelin
0.90
<0.01


H7C367
Non-POU domain-containing octamer-
1.02
0.03



binding protein


E9PLD1
Non-specific lipid-transfer protein
1.54
0.02


H0YFY6
Nuclear mitotic apparatus protein 1
1.10
0.02


Q02818
Nucleobindin-1
0.87
0.02


Q86U38
Nucleolar protein 9
0.86
0.03


E5RHP0
Nucleoside diphosphate kinase A
0.84
0.05


F8W543
Nucleosome assembly protein 1-like 1
0.75
0.02


P23515
Oligodendrocyte-myelin glycoprotein
0.80
<0.01


X6RKL2
Optineurin
0.81
<0.01


D6R9C5
Osteopontin
0.75
0.04


Q9BZF1
Oxysterol-binding protein-related
0.78
0.04



protein 8


Q96HC4
PDZ and LIM domain protein 5
0.75
0.02


Q9UBV8
Peflin
0.98
0.03


Q02790
Peptidyl-prolyl cis-trans isomerase
0.73
0.04



FKBP4


O14936
Peripheral plasma membrane protein
1.07
0.04



CASK


H7C5W5
Peripherin
1.14
0.04


P32119
Peroxiredoxin-2
0.96
0.03


I3L0T4
Peroxisomal acyl-coenzyme A oxidase 1
1.02
0.01


Q9Y285
Phenylalanine--tRNA ligase alpha
0.72
0.02



subunit


F8VVM2
Phosphate carrier protein,
0.78
0.01



mitochondrial


A8MYT4
Phosphatidylinositol 3-kinase
0.83
0.03


A8MTF1
Phosphatidylinositol 4-kinase alpha
0.71
0.01


P15259
Phosphoglycerate mutase 2
1.12
0.04


A6NDG6
Phosphoglycolate phosphatase
1.05
0.03


M0QZI4
Phospholipase D3
0.83
0.01


P36969
Phospholipid hydroperoxide
0.75
0.02



glutathione peroxidase, mitochondrial


Q9H008
Phospholysine phosphohistidine
0.74
0.03



inorganic pyrophosphate phosphatase


Q5SRE7
Phytanoyl-CoA dioxygenase domain-
0.65
0.01



containing protein 1


Q9GZP4
PITH domain-containing protein 1
0.71
0.04


Q504U3
PKM2 protein
0.73
0.02


I3L495
Platelet-activating factor
0.97
0.02



acetylhydrolase IB subunit alpha


Q09470
Potassium voltage-gated channel
0.73
0.04



subfamily A member 1


Q9UHV9
Prefoldin subunit 2
0.72
0.04


F8W8W4
Prenylcysteine oxidase 1
0.74
<0.01


Q8TBB6
Probable cationic amino acid
0.66
0.05



transporter


P01303
Pro-neuropeptide Y
0.86
0.04


Q9H7Z7
Prostaglandin E synthase 2
0.98
0.02


Q16186
Proteasomal ubiquitin receptor ADRM1
1.29
0.05


F5GX11
Proteasome subunit alpha type-1
0.84
0.03


P28066
Proteasome subunit alpha type-5
1.08
0.03


H0Y586
Proteasome subunit alpha type-7
0.78
0.03


P02760
Protein AMBP
−0.95
0.04


O60678
Protein arginine N-methyltransferase
1.27
0.01



3


Q5TA58
Protein argonaute
−0.78
0.03


E9PGA6
Protein C1QTNF3-AMACR
0.76
0.01


D6RAV0
Protein CDV3 homolog
0.69
0.01


B4DFG0
Protein DEK
0.84
0.01


I3L3P5
Protein disulfide-isomerase
0.86
0.01


Q92520
Protein FAM3C
0.68
0.03


Q13045
Protein flightless-1 homolog
0.65
<0.01


Q02156
Protein kinase C epsilon type
0.99
0.02


Q99435
Protein kinase C-binding protein
0.68
<0.01



NELL2


Q5SYT8
Protein NAMPTL
0.83
0.03


G3V2S0
Protein NDRG2
0.94
0.01


Q9BPW8
Protein NipSnap homolog 1
0.80
0.04


O75323
Protein NipSnap homolog 2
1.61
0.02


Q9UFN0
Protein NipSnap homolog 3A
0.73
0.04


G3V3Z8
Protein numb homolog
0.66
0.01


Q96A00
Protein phosphatase 1 regulatory
0.70
0.05



subunit 14A


Q9ULR3
Protein phosphatase 1H
0.85
0.01


Q9Y570
Protein phosphatase methylesterase 1
1.08
0.03


Q9Y6V0
Protein piccolo
0.93
0.04


E9PDC2
Protein prune homolog 2
1.12
0.03


K7EIR2
Protein QIL1
−0.74
0.03


P60903
Protein S100-A10
0.75
0.02


A8MRB1
Protein S100-B
1.05
0.02


H0Y8W8
Protein transport protein Sec31A
1.21
<0.01


H7BY58
Protein-L-isoaspartate O-
1.04
0.01



methyltransferase


Q5VT82
Protocadherin 9
1.02
<0.01


Q9P2E7
Protocadherin-10
0.78
0.01


A6NEC2
Puromycin-sensitive aminopeptidase-
0.69
0.04



like protein


P0C7P4
Putative cytochrome b-c1 complex
1.07
0.03



subunit Rieske-like protein 1


Q5VTE0
Putative elongation factor 1-alpha-
1.30
0.05



like 3


A8MUU1
Putative fatty acid-binding protein
0.80
0.02



5-like protein 3


Q6DN03
Putative histone H2B type 2-C
1.09
0.02


Q6GMV3
Putative peptidyl-tRNA hydrolase
0.67
0.01


Q9H853
Putative tubulin-like protein alpha-
0.69
0.04



4B


Q9NVS9
Pyridoxine-5′-phosphate oxidase
0.81
0.01


H3BTN5
Pyruvate kinase
0.71
0.03


E9PNP4
Radixin
3.05
0.04


Q96S59
Ran-binding protein 9
0.69
0.01


Q9Y4G8
Rap guanine nucleotide exchange
0.72
0.01



factor 2


Q13283
Ras GTPase-activating protein-binding
0.68
0.04



protein 1


P15153
Ras-related C3 botulinum toxin
1.05
0.04



substrate 2


P60763
Ras-related C3 botulinum toxin
0.97
0.03



substrate 3


B4DQU5
Ras-related protein Rab-11A
0.67
0.04


Q9UL25
Ras-related protein Rab-21
0.69
<0.01


K7ES41
Ras-related protein Rab-27B
1.46
0.01


Q8WUD1
Ras-related protein Rab-2B
0.80
0.01


Q9NP90
Ras-related protein Rab-9B
1.47
0.04


F6U784
Ras-related protein Rap-2a
1.16
0.05


O43353
Receptor-interacting
0.78
0.04



serine/threonine-protein kinase 2


P23471
Receptor-type tyrosine-protein
0.90
<0.01



phosphatase zeta


J3KQ66
Reelin
0.70
0.01


Q15493
Regucalcin
1.91
0.03


H0YLG5
Regulator of microtubule dynamics
0.93
0.04



protein 3


Q92900
Regulator of nonsense transcripts 1
0.80
0.03


B5MC59
Replication protein A 14 kDa subunit
0.71
<0.01


Q15293
Reticulocalbin-1
1.35
0.03


Q86UN3
Reticulon-4 receptor-like 2
1.18
0.01


Q5SYQ7
Retinal dehydrogenase 1
1.04
0.01


Q8TC12
Retinol dehydrogenase 11
−0.73
0.02


J3KRE2
Rho GDP-dissociation inhibitor 1
0.80
0.03


Q9P227
Rho GTPase-activating protein 23
0.88
<0.01


E9PMN0
Ribonuclease inhibitor
0.70
0.04


H0YB34
Ribonuclease UK114
1.04
0.02


Q9Y3A5
Ribosome maturation protein SBDS
0.86
0.05


O15034
RIMS-binding protein 2
2.48
0.04


Q5TZA2
Rootletin
0.77
0.03


H7C5W9
Sarcoplasmic/endoplasmic reticulum
0.77
0.05



calcium ATPase 2


D6RD99
Scrapie-responsive protein 1
0.70
0.02


P05060
Secretogranin-1
0.80
0.03


H0YKC2
Secretogranin-3
0.87
0.01


C9JDT0
Secretoneurin
1.16
0.01


O75326
Semaphorin-7A
0.85
<0.01


H7C299
Septin-5
0.83
0.02


K7EJ51
Septin-9
0.96
0.04


B4DLV4
Serine hydroxymethyltransferase
−1.13
0.03


P34897
Serine hydroxymethyltransferase,
−1.81
0.04



mitochondrial


O75494
Serine/arginine-rich splicing factor
0.79
0.02



10


E9PCD1
Serine/threonine-protein kinase WNK2
0.75
0.01


E9PH38
Serine/threonine-protein phosphatase
1.00
0.02



2A 65 kDa regulatory subunit A alpha



isoform


E9PHZ6
Serine/threonine-protein phosphatase
1.36
0.05



2A 65 kDa regulatory subunit A beta



isoform


Q68CR8
Serine/threonine-protein phosphatase
0.69
<0.01



2A activator


M0QWZ7
Serine--tRNA ligase, mitochondrial
1.17
0.02


Q6ZV89
SH2 domain-containing protein 5
−0.80
0.04


Q9BQI5
SH3-containing GRB2-like protein 3-
0.72
<0.01



interacting protein 1


P45954
Short/branched chain specific acyl-
1.11
<0.01



CoA dehydrogenase, mitochondrial


A6NMU3
Signal transducing adapter molecule 1
1.06
0.02


C9K0U8
Single-stranded DNA-binding protein,
0.67
0.04



mitochondrial


Q96PX8
SLIT and NTRK-like protein 1
0.77
0.02


K7EMD6
Small glutamine-rich
1.62
<0.01



tetratricopeptide repeat-containing



protein alpha


Q8NHG7
Small VCP/p97-interacting protein
0.76
0.03


Q99250
Sodium channel protein type 2 subunit
0.80
0.05



alpha


Q99884
Sodium-dependent proline transporter
1.26
0.01


Q5VZ42
Solute carrier family 12 member 5
0.83
<0.01


P61278
Somatostatin
1.03
0.03


Q9BX66
Sorbin and SH3 domain-containing
0.82
0.05



protein 1


Q99523
Sortilin
0.68
0.03


Q9H4F8
SPARC-related modular calcium-binding
0.91
<0.01



protein 1


H0YJE6
Spectrin beta chain, erythrocytic
0.80
<0.01


K7EJR2
Spermatogenesis-associated protein 22
0.66
0.03


H3BS51
Sphingomyelin phosphodiesterase 3
1.31
0.02


Q13838
Spliceosome RNA helicase DDX39B
0.85
0.05


H0Y9U2
Splicing factor, proline- and
1.45
0.01



glutamine-rich


Q9HCB6
Spondin-1
0.75
<0.01


A2A2D0
Stathmin
0.78
0.04


P31040
Succinate dehydrogenase [ubiquinone]
1.05
0.03



flavoprotein subunit, mitochondrial


C9J8Q5
Succinate-semialdehyde dehydrogenase,
0.77
0.01



mitochondrial


P17600
Synapsin-1
0.95
0.03


Q92777
Synapsin-2
0.86
0.03


Q496J9
Synaptic vesicle glycoprotein 2C
1.15
0.02


K7EM19
Synaptic vesicle membrane protein
0.78
0.04



VAT-1 homolog


H7C4W3
Synaptophysin
2.38
0.03


C9J0A2
Synaptoporin
0.92
0.03


F5GX00
Synaptotagmin-7
1.50
<0.01


F5GZI8
T-complex protein 1 subunit alpha
0.89
0.03


P78371
T-complex protein 1 subunit beta
0.69
0.02


E9PM09
T-complex protein 1 subunit gamma
1.64
0.03


P24821
Tenascin
0.73
0.05


Q08629
Testican-1
1.73
<0.01


Q9BQ16
Testican-3
0.84
0.03


J3QL04
Tether-containing UBX domain for
1.13
0.03



GLUT4


H0YB37
Tetratricopeptide repeat protein 1
1.03
0.02


Q86TV6
Tetratricopeptide repeat protein 7B
0.95
<0.01


P10599
Thioredoxin
0.93
0.03


K7EME7
Thioredoxin-like protein 1
1.28
0.01


C9JV37
Thrombin light chain
−0.71
0.03


Q9Y2W1
Thyroid hormone receptor-associated
1.19
0.03



protein 3


G3V1L9
Tight junction protein 1 (Zona
0.67
0.03



occludens 1), isoform CRA_a


P13726
Tissue factor
0.68
0.02


E7EN89
Toll interacting protein, isoform
0.74
0.01



CRA_b


F2Z393
Transaldolase
0.86
0.01


Q5H9L2
Transcription elongation factor A
1.76
0.01



protein-like 5


E9PL10
Transcription factor BTF3 homolog 4
1.33
<0.01


M0R3C0
Transcription intermediary factor 1-
0.78
0.04



beta


Q00577
Transcriptional activator protein
1.06
0.04



Pur-alpha


B4DQI6
Transformer-2 protein homolog alpha
0.77
0.02


H7BXF3
Transformer-2 protein homolog beta
0.72
0.04


J3KQ45
Trans-Golgi network integral membrane
0.80
0.05



protein 2


F8W888
Transketolase
1.44
<0.01


Q9BSH4
Translational activator of cytochrome
1.13
0.01



c oxidase 1 (Coiled-coil domain-



containing protein 44) (Translational



activator of mitochondrially-encoded



cytochrome c oxidase I)


Q53FP2
Transmembrane protein 35
0.87
0.05


Q9BTV4
Transmembrane protein 43
−0.80
0.05


C9JE81
Trifunctional enzyme subunit beta,
0.68
0.02



mitochondrial


P60174
Triosephosphate isomerase
1.00
0.05


F2Z2W7
tRNA (uracil-5-)-methyltransferase
0.89
<0.01



homolog A


Q9Y3I0
tRNA-splicing ligase RtcB homolog
1.15
0.01


P28289
Tropomodulin-1
1.00
0.04


Q15714
TSC22 domain family protein 1
0.89
<0.01


F8VXZ7
Tubulin alpha-1A chain
0.95
0.01


M0QZL7
Tubulin beta-4A chain
0.75
0.01


D6RG15
Twinfilin-2
−0.99
0.01


P06241
Tyrosine-protein kinase Fyn (EC
0.79
0.02



2.7.10.2) (Proto-oncogene Syn)



(Proto-oncogene c-Fyn) (Src-like



kinase) (SLK) (p59-Fyn)


E9PLZ4
Tyrosine-protein phosphatase non-
0.98
0.02



receptor type 5


M0QYR1
U1 small nuclear ribonucleoprotein 70
0.75
<0.01



kDa (Fragment)


Q9UHD9
Ubiquilin-2
1.15
0.01


K7EJ02
UBX domain-containing protein 6
−0.78
0.01


Q14376
UDP-glucose 4-epimerase
0.82
0.02


E9PPU6
Uncharacterized protein
−0.70
0.01


Q9BXV9
Uncharacterized protein C14orf142
0.70
0.03


H0YEV9
Unconventional myosin-XVIIIa
−0.81
0.02


Q9H3H3
UPF0696 protein C11orf68
0.83
0.01


K7ELW1
UV excision repair protein RAD23
0.81
0.01



homolog A


Q5W0S4
UV excision repair protein RAD23
0.96
<0.01



homolog B


Q709C8
Vacuolar protein sorting-associated
1.45
0.03



protein 13C


Q6EMK4
Vasorin
0.80
0.04


P49748
Very long-chain specific acyl-CoA
0.91
0.01



dehydrogenase, mitochondrial


B0YJC4
Vimentin
0.80
0.03


H0Y715
Voltage-dependent calcium channel
0.87
0.01



subunit alpha-2/delta-1


B5MCX6
V-set and transmembrane domain-
0.81
0.01



containing protein 2A


K7ERA0
V-type proton ATPase subunit a
0.74
<0.01


Q8N8Y2
V-type proton ATPase subunit d 2
1.66
0.01


Q15904
V-type proton ATPase subunit S1
1.00
0.02


Q8TF74
WAS/WASL-interacting protein family
1.71
0.02



member 2


H0YMF9
WD repeat-containing protein 61
−2.20
<0.01


Q9UPY6
Wiskott-Aldrich syndrome protein
0.77
0.03



family member 3


P61129
Zinc finger CCCH domain-containing
1.08
0.04



protein 6


Q9ULF5
Zinc transporter ZIP10
0.76
0.02









In one embodiment, the panel of biomarkers comprises at least Basigin.


In another embodiment, the panel of biomarkers comprises at least Cytochrome c oxidase subunit 7A-related protein, mitochondrial.


In another embodiment, the panel of biomarkers comprises at least Basigin and Cytochrome c oxidase subunit 7A-related protein, mitochondrial.


Basigin or BSG or extracellular matrix metalloproteinase inducer (EMMPRIN) or cluster of differentiation 147 (CD147) is a type I integral membrane receptor that has many ligands, including the cyclophilin (CyP) proteins Cyp-A and CyP-B and certain integrins. Basigin has metalloproteinase-inducing ability and it also regulates several distinct functions, such as spermatogenesis, expression of the monocarboxylate transporter and the responsiveness of lymphocytes. It is expressed by many cell types, including epithelial cells, endothelial cells and leukocytes. The human Basigin protein contains 269 amino acids that form two heavily glycosylated C2 type immunoglobulin-like domains at the N-terminal extracellular portion. A second form of Basigin has also been characterized that contains one additional immunoglobulin-like domain in its extracellular portion. Its amino acid sequence is depicted in SEQ ID NO: 30.


Cytochrome c oxidase subunit 7A-related protein, mitochondrial or Cox7r is an enzyme encoded by the COX7A2L gene in humans. Cytochrome c oxidase subunit 7A-related protein, mitochondrial is a component of the Cytochrome c oxidase (COX), which is the terminal component of the mitochondrial respiratory chain and catalyzes the electron transfer from reduced cytochrome c to oxygen. Its amino acid sequence is depicted in SEQ ID NO: 31.


The embodiments relating to the biomarkers selected from Table 5, as described above, are equally applicable to all other embodiments of the third aspect of the invention and to all other aspects of the invention where biomarkers selected from Table 5 are involved.


In one embodiment, the panel of biomarker comprises at least one biomarker selected from Table 5 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one biomarker selected from Groups A, B, C or D. More preferably the biomarker selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


The biomarkers listed in Table 5 may belong to specific pathways which are known to be relevant in the pathology of AD. They may be further grouped in specific lists as those illustrated below in Tables 6 to 13 according to specific GO terms (http://geneontology.org/).


One GO term used to analyse the data was the term “Synap*”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 6.


In one embodiment, the panel of biomarkers according to the invention comprises one or more, preferably two or more biomarkers selected from Table 6.









TABLE 6







Proteins with GO Term ‘Synap*’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













P05067
Amyloid beta A4 protein
0.90
<0.01


Q9UQM7
Calcium/calmodulin-dependent protein
0.81
0.02



kinase type II subunit alpha


Q16566
Calcium/calmodulin-dependent protein
0.85
0.03



kinase type IV


C9IZ88
Catenin alpha-2
2.11
0.01


Q5JYX0
Cell division control protein 42 homolog
1.04
0.05


I3L0M4
Coiled-coil domain-containing protein 43
0.99
0.01


Q8NFT8
Delta and Notch-like epidermal growth
1.02
0.01



factor-related receptor


P50570
Dynamin-2
0.89
0.03


P42261
Glutamate receptor 1
1.13
0.04


P01112
GTPase HRas
1.09
0.01


M0R2G0
Leucine-rich repeat-containing protein 4B
0.76
<0.01


Q9Y4C0
Neurexin-3
0.93
<0.01


Q9HDB5
Neurexin-3-beta
0.98
<0.01


P61601
Neurocalcin-delta
1.33
0.03


Q9UBB6
Neurochondrin
0.80
0.02


Q8NFZ4
Neuroligin-2
0.96
0.04


P47972
Neuronal pentraxin-2
1.02
0.01


Q96HC4
PDZ and LIM domain protein 5
0.75
0.02


I3L495
Platelet-activating factor
0.97
0.02



acetylhydrolase IB subunit alpha


Q09470
Potassium voltage-gated channel subfamily
0.73
0.04



A member 1


P01303
Pro-neuropeptide Y
0.86
0.04


Q02156
Protein kinase C epsilon type
0.99
0.02


Q9Y6V0
Protein piccolo
0.93
0.04


Q9Y4G8
Rap guanine nucleotide exchange factor 2
0.72
0.01


P61278
Somatostatin
1.03
0.03


P17600
Synapsin-1
0.95
0.03


Q92777
Synapsin-2
0.86
0.03


H7C4W3
Synaptophysin
2.38
0.03









In one embodiment, the panel of biomarker comprises at least one biomarker selected from Table 6 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one biomarker selected from Groups A, B, C or D. More preferably the biomarker selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Another GO term used to analyse the data was the term “Phosphoryl*”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 7.


In another embodiment, the panel of biomarkers according to the invention comprises one or more, preferably two or more biomarkers selected from Table 7.









TABLE 7







Proteins with GO Term ‘Phosporyl*’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













Q8IZP0
Abl interactor 1
1.04
0.01


P05067
Amyloid beta A4 protein
0.90
<0.01


P03950
Angiogenin
−0.76
0.03


P17858
ATP-dependent 6-phosphofructokinase,
0.68
0.02



liver type


P11274
Breakpoint cluster region protein
0.99
0.01


Q9UQM7
Calcium/calmodulin-dependent protein
0.81
0.02



kinase type II subunit alpha


Q16566
Calcium/calmodulin-dependent protein
0.85
0.03



kinase type IV


P31321
cAMP-dependent protein kinase type I-
1.56
0.04



beta regulatory subunit


Q5JYX0
Cell division control protein 42
1.04
0.05



homolog


O75367
Core histone macro-H2A.1
0.67
0.01


P14927
Cytochrome b-c1 complex subunit 7
0.98
0.05


Q08495
Dematin
0.76
0.01


P20827
Ephrin-A1
0.86
0.05


E5RGR6
GDNF family receptor alpha-2
1.06
<0.01


P01112
GTPase HRas
1.09
0.01


H0YB38
Inositol monophosphatase 3
1.12
0.04


K7EKJ9
Interleukin enhancer-binding factor 3
1.57
0.04


A2A2V1
Major prion protein
1.20
<0.01


Q15746
Myosin light chain kinase, smooth
1.08
0.01



muscle


O14936
Peripheral plasma membrane protein
1.07
0.04



CASK


A8MTF1
Phosphatidylinositol 4-kinase alpha
0.71
0.01


A6NDG6
Phosphoglycolate phosphatase
1.05
0.03


Q9H008
Phospholysine phosphohistidine
0.74
0.03



inorganic pyrophosphate phosphatase


Q02156
Protein kinase C epsilon type
0.99
0.02


Q5SYT8
Protein NAMPTL
0.83
0.03


O75323
Protein NipSnap homolog 2
1.61
0.02


Q96A00
Protein phosphatase 1 regulatory
0.70
0.05



subunit 14A


O43353
Receptor-interacting serine/threonine-
0.78
0.04



protein kinase 2


P23471
Receptor-type tyrosine-protein
0.90
<0.01



phosphatase zeta


O75326
Semaphorin-7A
0.85
<0.01


M0R3C0
Transcription intermediary factor 1-
0.78
0.04



beta


P06241
Tyrosine-protein kinase Fyn (EC
0.79
0.02



2.7.10.2) (Proto-oncogene Syn) (Proto-



oncogene c-Fyn) (Src-like kinase)



(SLK) (p59-Fyn)









In one embodiment, the panel of biomarker comprises at least one biomarker selected from Table 7 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one biomarker selected from Groups A, B, C or D. More preferably the biomarker selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Another GO term used to analyse the data was the term “Stress”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 8.


In another embodiment, the panel of biomarkers according to the invention comprises one or more, preferably two or more biomarkers selected from Table 8.









TABLE 8







Proteins with GO Term ‘Stress’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













P11766
Alcohol dehydrogenase class-3
0.87
0.05


P05067
Amyloid beta A4 protein
0.90
<0.01


E9PMA0
Apoptosis-inducing factor 1,
0.69
0.03



mitochondrial


P29972
Aquaporin-1
0.90
<0.01


Q99674
Cell growth regulator with EF hand
0.86
<0.01



domain protein 1


H0YIV0
Endoplasmin
1.18
0.04


P07203
Glutathione peroxidase 1
1.13
0.02


C9J3N8
Heat shock protein beta-1
−0.91
0.01


Q9Y4L1
Hypoxia up-regulated protein 1
0.86
0.02


Q13136
Liprin-alpha-1
0.85
0.01


A2A2V1
Major prion protein
1.20
<0.01


E5RJK1
Mitochondrial peptide methionine
0.87
<0.01



sulfoxide reductase


P32119
Peroxiredoxin-2
0.96
0.03


P60903
Protein S100-A10
0.75
0.02


O43353
Receptor-interacting serine/threonine-
0.78
0.04



protein kinase 2


Q99250
Sodium channel protein type 2 subunit
0.80
0.05



alpha


Q9BX66
Sorbin and SH3 domain-containing
0.82
0.05



protein 1









In yet another embodiment, the panel of biomarker comprises at least one biomarker selected from Table 8 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one or more biomarker selected from Groups A, B, C or D. More preferably the one or more biomarkers selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Another GO term used to analyse the data was the term “Calcium”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 9.


In another embodiment, the panel of biomarkers according to the invention comprises one or more, preferably two or more biomarkers selected from Table 9.









TABLE 9







Proteins with GO Term ‘Calcium’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













P05067
Amyloid beta A4 protein
0.90
<0.01


E7EV01
Ankyrin repeat and SOCS box protein 2
1.68
0.01


P07686
Beta-hexosaminidase subunit beta
1.49
<0.01


P19022
Cadherin-2
1.17
0.05


Q9UQM7
Calcium/calmodulin-dependent protein
0.81
0.02



kinase type II subunit alpha


P07384
Calpain-1 catalytic subunit
1.06
0.03


Q96A33
Coiled-coil domain-containing protein
0.79
0.02



47


Q08495
Dematin
0.76
0.01


P78417
Glutathione S-transferase omega-1
1.13
0.01


P13645
Keratin, type I cytoskeletal 10
0.66
0.02


P01042
Kininogen-1
−0.67
0.02


Q15746
Myosin light chain kinase, smooth
1.08
0.01



muscle


P61601
Neurocalcin-delta
1.33
0.03


Q9UBV8
Peflin
0.98
0.03


O14936
Peripheral plasma membrane protein
1.07
0.04



CASK


P01303
Pro-neuropeptide Y
0.86
0.04


Q02156
Protein kinase C epsilon type
0.99
0.02


Q15493
Regucalcin
1.91
0.03


H7C5W9
Sarcoplasmic/endoplasmic reticulum
0.77
0.05



calcium ATPase 2


P06241
Tyrosine-protein kinase Fyn (EC
0.79
0.02



2.7.10.2) (Proto-oncogene Syn) (Proto-



oncogene c-Fyn) (Src-like kinase)



(SLK) (p59-Fyn)









In one another embodiment, the panel of biomarker comprises at least one biomarker selected from Table 9 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one biomarker selected from Groups A, B, C or D. More preferably the biomarker selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Another GO term used to analyse the data was the term “Cytoskelet*”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 10.


In another embodiment, the panel of biomarker according to the invention comprises one or more, preferably two or more biomarkers selected from Table 10.









TABLE 10







Proteins with GO Term ‘Cytoskelet*’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













Q8IZP0
Abl interactor 1
1.04
0.01


E9PF58
Actin-related protein 2/3 complex
0.94
0.02



subunit 1A


P16157
Ankyrin-1
0.86
0.01


P04114
Apolipoprotein B-100
0.70
0.04


Q4VXN1
Band 4.1-like protein 1
1.36
0.01


Q562R1
Beta-actin-like protein 2
0.77
0.05


C9IZ88
Catenin alpha-2
2.11
0.01


Q14019
Coactosin-like protein
0.68
0.02


Q08495
Dematin
0.76
0.01


P60981
Destrin
0.93
0.05


Q15334
Lethal(2) giant larvae protein homolog
0.74
0.04



1


O14745
Na(+)/H(+) exchange regulatory
1.06
0.05



cofactor NHE-RF1


Q96HC4
PDZ and LIM domain protein 5
0.75
0.02


O14936
Peripheral plasma membrane protein
1.07
0.04



CASK


Q02156
Protein kinase C epsilon type
0.99
0.02


Q9Y6V0
Protein piccolo
0.93
0.04


O43353
Receptor-interacting serine/threonine-
0.78
0.04



protein kinase 2


Q5TZA2
Rootletin
0.77
0.03


B4DLV4
Serine hydroxymethyltransferase
−1.13
0.03


P34897
Serine hydroxymethyltransferase,
−1.81
0.04



mitochondrial


P28289
Tropomodulin-1
1.00
0.04


B0YJC4
Vimentin
0.80
0.03


Q8TF74
WAS/WASL-interacting protein family
1.71
0.02



member 2


Q9UPY6
Wiskott-Aldrich syndrome protein
0.77
0.03



family member 3









In another embodiment, the panel of biomarker comprises at least one biomarker selected from Table 10 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one or more biomarkers selected from Groups A, B, C or D. More preferably the one or more biomarkers selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


Yet another GO term used to analyse the data was the term “Mitochondri*”. Biomarkers of Table 5 which are registered as part of this specific pathway are shown in Table 11.


In another embodiment, the panel of biomarkers according to the invention comprises one or more, preferably two or more biomarkers selected from Table 11.









TABLE 11







Proteins with GO Term ‘Mitochondri*’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













K7EJ68
3-ketoacyl-CoA thiolase, mitochondrial
0.91
0.01


P25325
3-mercaptopyruvate sulfurtransferase
0.81
0.01


O14561
Acyl carrier protein, mitochondrial
0.72
0.02


P07108
Acyl-CoA-binding protein
1.18
0.02


O14734
Acyl-coenzyme A thioesterase 8
0.76
<0.01


P12235
ADP/ATP translocase 1
0.71
0.03


P11766
Alcohol dehydrogenase class-3
0.87
0.05


E9PMA0
Apoptosis-inducing factor 1,
0.69
0.03



mitochondrial


O43776
Asparagine--tRNA ligase, cytoplasmic
1.37
0.03


K7EJP1
ATP synthase subunit alpha,
0.77
0.04



mitochondrial


Q86VU5
Catechol O-methyltransferase domain-
1.06
0.01



containing protein 1


Q8NI60
Chaperone activity of bc1 complex-like,
0.78
0.01



mitochondrial


H3BP04
Cytochrome b-c1 complex subunit 2,
0.67
0.02



mitochondrial


P14927
Cytochrome b-c1 complex subunit 7
0.98
0.05


P47985
Cytochrome b-c1 complex subunit Rieske,
1.13
0.04



mitochondrial


Q5JTJ3
Cytochrome c oxidase assembly factor 6
1.23
0.01



homolog


K7EQD3
Cytochrome c oxidase subunit 6B1
0.88
0.02


E5RJZ1
Cytochrome c oxidase subunit 7A-related
4.19
0.05



protein, mitochondrial


Q9NVH1
DnaJ homolog subfamily C member 11
0.94
0.01


Q92506
Estradiol 17-beta-dehydrogenase 8
0.87
0.04


Q92731
Estrogen receptor beta
1.73
0.01


G3V1D1
Ferritin
1.37
0.03


P07203
Glutathione peroxidase 1
1.13
0.02


P23434
Glycine cleavage system H protein,
0.94
0.01



mitochondrial


P08107
Heat shock 70 kDa protein 1A/1B
0.70
0.03


F5GWX2
Heme-binding protein 1
1.24
<0.01


B1AR61
Hexokinase-1
0.83
0.02


Q16775
Hydroxyacylglutathione hydrolase,
0.85
0.04



mitochondrial


K7EKJ9
Interleukin enhancer-binding factor 3
1.57
0.04


Q5VVL7
Lipoamide acyltransferase component of
0.84
0.03



branched-chain alpha-keto acid



dehydrogenase complex, mitochondrial


A8MW50
L-lactate dehydrogenase
0.72
0.02


F5GZQ4
L-lactate dehydrogenase A chain
0.68
<0.01


O95573
Long-chain-fatty-acid--CoA ligase 3
0.82
0.01


C9JF79
Malate dehydrogenase
1.07
0.04


Q5HYI7
Metaxin-3
1.07
<0.01


Q13825
Methylglutaconyl-CoA hydratase,
1.50
0.03



mitochondrial


Q9H936
Mitochondrial glutamate carrier 1
0.66
<0.01


G3V502
Mitochondrial import inner membrane
0.92
0.03



translocase subunit Tim9


E5RJK1
Mitochondrial peptide methionine
0.87
<0.01



sulfoxide reductase


Q10713
Mitochondrial-processing peptidase
0.70
0.04



subunit alpha


D6RAU3
Mitogen-activated protein kinase 10
0.65
0.04


O95865
N(G),N(G)-dimethylarginine
0.83
0.01



dimethylaminohydrolase 2


Q4G0N4
NAD kinase 2, mitochondrial
0.96
<0.01


D6RAI5
NAD(P) transhydrogenase, mitochondrial
0.86
<0.01


F8VRD8
NADH dehydrogenase [ubiquinone] 1 alpha
1.00
0.05



subcomplex subunit 12


O95182
NADH dehydrogenase [ubiquinone] 1 alpha
0.73
0.02



subcomplex subunit 7


Q16795
NADH dehydrogenase [ubiquinone] 1 alpha
0.74
0.04



subcomplex subunit 9, mitochondrial


E9PQ68
NADH dehydrogenase [ubiquinone] 1 beta
−0.85
<0.01



subcomplex subunit 8, mitochondrial


P56181
NADH dehydrogenase [ubiquinone]
0.70
0.03



flavoprotein 3, mitochondrial


Q9UHQ9
NADH-cytochrome b5 reductase 1
0.72
0.02


Q02790
Peptidyl-prolyl cis-trans isomerase
0.73
0.04



FKBP4


F8VVM2
Phosphate carrier protein, mitochondrial
0.78
0.01


P36969
Phospholipid hydroperoxide glutathione
0.75
0.02



peroxidase, mitochondrial


Q9UHV9
Prefoldin subunit 2
0.72
0.04


Q9H7Z7
Prostaglandin E synthase 2
0.98
0.02


Q02156
Protein kinase C epsilon type
0.99
0.02


Q9BPW8
Protein NipSnap homolog 1
0.80
0.04


O75323
Protein NipSnap homolog 2
1.61
0.02


K7EIR2
Protein QIL1
−0.74
0.03


H3BTN5
Pyruvate kinase
0.71
0.03


H0YLG5
Regulator of microtubule dynamics
0.93
0.04



protein 3


B4DLV4
Serine hydroxymethyltransferase
−1.13
0.03


P34897
Serine hydroxymethyltransferase,
−1.81
0.04



mitochondrial


P45954
Short/branched chain specific acyl-CoA
1.11
<0.01



dehydrogenase, mitochondrial


C9K0U8
Single-stranded DNA-binding protein,
0.67
0.04



mitochondrial


P31040
Succinate dehydrogenase [ubiquinone]
1.05
0.03



flavoprotein subunit, mitochondrial


C9J8Q5
Succinate-semialdehyde dehydrogenase,
0.77
0.01



mitochondrial


P10599
Thioredoxin
0.93
0.03


Q9BSH4
Translational activator of cytochrome c
1.13
0.01



oxidase 1 (Coiled-coil domain-containing



protein 44) (Translational activator of



mitochondrially-encoded cytochrome c



oxidase I)


C9JE81
Trifunctional enzyme subunit beta,
0.68
0.02



mitochondrial


P06241
Tyrosine-protein kinase Fyn (EC
0.79
0.02



2.7.10.2) (Proto-oncogene Syn) (Proto-



oncogene c-Fyn) (Src-like kinase) (SLK)



(p59-Fyn)


Q6EMK4
Vasorin
0.80
0.04


P49748
Very long-chain specific acyl-CoA
0.91
0.01



dehydrogenase, mitochondrial









In another embodiment, the panel of biomarker comprises at least one biomarker selected from Table 11 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one or more biomarkers selected from Groups A, B, C or D. More preferably the one or more biomarkers selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


The biomarkers of Table 5 were also analysed with the GO terms “Vesicle” and “Insuline”. Those which are registered as part of this specific pathways are shown in Table 12 and Table 13, respectively.


In another embodiment, the panel of biomarker according to the invention comprises one or more, preferably two or more biomarkers selected from Table 12 or Table 13.









TABLE 12







Proteins with GO Term ‘Vesicle’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













P16157
Ankyrin-1
0.86
0.01


B0YIW6
Archain 1
−1.26
0.01


Q9UPT6
C-Jun-amino-terminal kinase-
−0.85
0.01



interacting protein 3


D6RJD1
Clathrin light chain B
0.84
0.02


P53618
Coatomer subunit beta
1.07
0.03


H0Y8X7
Coatomer subunit gamma-1
0.69
0.03


P50570
Dynamin-2
0.89
0.03


P61601
Neurocalcin-delta
1.33
0.03


I3L495
Platelet-activating factor
0.97
0.02



acetylhydrolase IB subunit alpha


Q9Y6V0
Protein piccolo
0.93
0.04


Q8WUD1
Ras-related protein Rab-2B
0.80
0.01


Q99523
Sortilin
0.68
0.03


P17600
Synapsin-1
0.95
0.03
















TABLE 13







Proteins with GO Term ‘Insulin’ found


to be regulated in CSF of AD patients












Log2



UniProtKB

AD/


Accession

Con-
P


Number
Protein name
trol
value













P12235
ADP/ATP translocase 1
0.71
0.03


P17174
Aspartate aminotransferase,
0.99
0.03



cytoplasmic


P17858
ATP-dependent 6-phosphofructokinase,
0.68
0.02



liver type


P31321
cAMP-dependent protein kinase type I-
1.56
0.04



beta regulatory subunit


Q9UKG1
DCC-interacting protein 13-alpha
0.79
0.01


P01112
GTPase HRas
1.09
0.01


P63092
Guanine nucleotide-binding protein
1.00
0.03



G(s) subunit alpha isoforms short


P16519
Neuroendocrine convertase 2
1.52
0.00


O15240
Neurosecretory protein VGF
1.12
0.00


Q9BZF1
Oxysterol-binding protein-related
0.78
0.04



protein 8


Q02156
Protein kinase C epsilon type
0.99
0.02


Q9Y6V0
Protein piccolo
0.93
0.04


Q9BX66
Sorbin and SH3 domain-containing
0.82
0.05



protein 1


Q99523
Sortilin
0.68
0.03


Q8N8Y2
V-type proton ATPase subunit d 2
1.66
0.01









In other embodiments, the panel of biomarker comprises at least one biomarker selected from Table 12 or Table 13 and at least one biomarker selected from tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and
    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid.


Preferably, the panel also comprises one or more biomarkers selected from Groups A, B, C or D. More preferably the one or more biomarkers selected from Groups A, B, C or D is:

    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;
    • or
    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof.


All non-modified peptides for each protein were summed for three control and three AD cases. The log 2 ratio and p-value of each protein was then calculated. Proteins with 2 or more peptides, >60% regulation and p<0.05 were selected as biomarkers of synaptic dysfunction (Table 6), dysregulated phosphorylation (Table 7), oxidative stress (Table 8), dysregulated calcium signalling (Table 9), dysregulated cytoskeleton (Table 10), mitochondrial damage (Table 11), abnormal vesicle function (Table 12) and dysfunctional insulin signalling (Table 13).


The panels of biomarkers described herein are useful for diagnosing, for staging, for assessing the likelihood of developing a neurocognitive disorder, and for assessing the response to a drug for treating a neurocognitive disorder, such as a neurocognitive disease characterized by tau toxicity, for example a tauopathy and in particular Alzheimer's disease. The use of these panels of biomarkers according to the present invention in any of such methods has considerable advantages.


Firstly, the biomarkers according to the present invention represent the translation of tau toxicity and resulting changes in pathways that occur in the brain into a peripheral signal in a peripheral tissue such as CSF and blood. Hence, they allow replacing tissue testing with a peripheral fluid testing. This represents a great advantage especially as the tissue primarily affected in neurocognitive disorder is the brain tissue. Brain biopsies are not carried out unless post-mortem.


Secondly, the biomarkers according to the present invention have been selected as those being capable to translate the specific stage of a neurocognitive disorder characterized by tau toxicity, such as Alzheimer's disease. This also represents a great advantage as currently clinicians assess the advancement of a neurocognitive diseases like AD through a battery of psychometric tests, which, albeit being somewhat indicative of the disease progression, may not be precise on the stage of the disease, thus, making it particularly difficult to select those therapies which have been developed and approved for a specific stage.


Thirdly, these biomarkers further comprise proteins which are not those typically reported in the literature or currently used in the clinical setting as biomarkers for tauopathies, thus providing clinicians with additional tools for identifying and distinguishing, even at an early stage, subjects who have a neurocognitive disorder characterized by tau toxicity, such as AD and subjects who, despite presenting symptoms of neurocognitive impairment are not affected by the early signs of AD.


Hence, the present invention provides for a method for diagnosing a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for biomarkers of a panel as defined herein;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers of the panel in said sample with reference concentrations or amounts of said biomarkers;


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





Preferably, the neurocognitive disorder is characterized by tau-toxicity; more preferably the neurocognitive disorder is a tauopathy selected from the group of Alzheimer's disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy (PSP), Pick's disease, corticobasal degeneration, multisystem atrophy (MSA), neurobasal degeneration with iron accumulation, type 1 (Hallervorden-Spatz), argyrophilic grain dementia, Down's syndrome, diffuse neurofibrillary tangles with calcification, dementia pugilistica, Gerstmann-Straussler-Scheinker disease, myotonic dystrophy, Niemann-Pick disease type C, progressive subcortical gliosis, prion protein cerebral amyloid angiopathy, tangle only dementia, postencephalitic parkinsonism, subacute sclerosing panencephalitis, Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex, non-Guamanian motor neuron disease with neurofibrillary tangles/dementia, chronic traumatic encephalopathy, alpha-synucleinopathies, Parkinson's disease or combinations thereof.


Even more preferably the tauopathy is Alzheimer's disease.


The present invention also provide for a method for staging a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for biomarkers of a panel as defined herein;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining the stage of the neurocognitive disorder in said subject by comparing said concentration or amount of each of the biomarkers of the panel in said sample with reference concentrations or amounts of said biomarkers;


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





Preferably, in the staging method according to the invention, the levels of protein phosphatase 1 regulatory subunit 14A are increased in said sample of a subject with an advanced stage of the neurocognitive disorder; and/or the levels of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase are increased in said sample of a subject with an advanced stage of the neurocognitive disorder. In one embodiment, the staging of the neurocognitive disorder is higher (i.e. more advanced stage) the higher the tau expression and hyperphosphorylation. Hence, the stage is tau-dependent.


Preferably, the neurocognitive disorder is characterized by tau-toxicity and more preferably the neurocognitive disorder is a tauopathy selected from the group of Alzheimer's disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy (PSP), Pick's disease, corticobasal degeneration, multisystem atrophy (MSA), neurobasal degeneration with iron accumulation, type 1 (Hallervorden-Spatz), argyrophilic grain dementia, Down's syndrome, diffuse neurofibrillary tangles with calcification, dementia pugilistica, Gerstmann-Straussler-Scheinker disease, myotonic dystrophy, Niemann-Pick disease type C, progressive subcortical gliosis, prion protein cerebral amyloid angiopathy, tangle only dementia, postencephalitic parkinsonism, subacute sclerosing panencephalitis, Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex, non-Guamanian motor neuron disease with neurofibrillary tangles/dementia, chronic traumatic encephalopathy, alpha-synucleinopathies, Parkinson's disease or combinations thereof.


Even more preferably the tauopathy is Alzheimer's disease (AD) and the staging is any one of the stages of Braak staging of AD.


Braak staging for AD was firstly described in 1991 (Braak, H. et al. (1991) Acta Neuropathologica 82 (4): 239-59) and comprises:

    • stages I and II: used when neurofibrillary tangle involvement is confined mainly to the transentorhinal region of the brain;
    • stages III and IV when there is also involvement of limbic regions such as the hippocampus, and
    • stages V and VI when there is extensive neocortical involvement


In one preferred embodiment, when the the neurocognitive disorder is AD, the concentration or amount of protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof is increased in the sample of AD patients with AD Braak stage V or Braak stage VI with respect to AD patients with AD Braak stage III or stage IV.


In another preferred embodiment, when the the neurocognitive disorder is AD, the concentration or amount of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof is increased in the sample of AD patients with AD Braak stage V or Braak stage VI with respect to AD patients with AD Braak stage III or stage IV.


The invention also provides a method for assessing in a subject the likelihood of developing a neurocognitive disorder, the method comprising:

    • a) assaying a sample obtained from said subject for biomarkers of a panel as defined herein;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject is likely to develop a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers of the biomarker panel in said sample with reference concentrations or amounts of said biomarkers;


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





Preferably, the neurocognitive disorder is characterized by tau-toxicity and more preferably the neurocognitive disorder is a tauopathy selected from the group of Alzheimer's disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy (PSP), Pick's disease, corticobasal degeneration, multisystem atrophy (MSA), neurobasal degeneration with iron accumulation, type 1 (Hallervorden-Spatz), argyrophilic grain dementia, Down's syndrome, diffuse neurofibrillary tangles with calcification, dementia pugilistica, Gerstmann-Straussler-Scheinker disease, myotonic dystrophy, Niemann-Pick disease type C, progressive subcortical gliosis, prion protein cerebral amyloid angiopathy, tangle only dementia, postencephalitic parkinsonism, subacute sclerosing panencephalitis, Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex, non-Guamanian motor neuron disease with neurofibrillary tangles/dementia, chronic traumatic encephalopathy, alpha-synucleinopathies, Parkinson's disease or combinations thereof.


Even more preferably the tauopathy is Alzheimer's disease.


The present invention also provides for a method for treating a neurocognitive disorder in a subject, the method comprising:

    • a) assaying a sample obtained from said subject for biomarkers of a panel as defined herein;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers;
    • d) administering to said subject a drug for treating the neurocognitive disorder;


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





Alternatively, this aspect may be formulated as a drug for use in the treatment of a neurocognitive disorder in a subject, wherein the subject is selected by the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers; wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.


      or, alternatively,


      as a use of a drug for the manufacture of a medicament for the treatment of a neurocognitive disorder in a subject, wherein the subject is selected by the method comprising:

    • a) assaying a sample obtained from said subject for the biomarkers of the panel as defined in any one of the first, second and third aspect of the invention, including the embodiments thereof;

    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said panel;

    • c) determining whether that subject has a neurocognitive disorder by comparing said concentration or amount of each of the biomarkers in said sample with reference concentrations or amounts of said biomarkers; wherein the panel of biomarkers is selected from a panel comprising:





I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





In one embodiment, when the sample is a brain sample, the concentration or amount of protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof will decrease in response to the administration of the drug for treating the neurocognitive disorder.


In another embodiment, when the sample is CSF, the concentration or amount of protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform or a variant or a fragment thereof increase or decrease in response to the treatment.


The present invention also provide for a method for assessing the response to a drug for treating a neurocognitive disorder in a subject, wherein the subject has been treated or is being treated with said drug, the method comprises:

    • a) assaying a sample obtained from said subject for biomarkers of a panel as described herein;
    • b) measuring in said sample a concentration or an amount of each of the biomarkers of said biomarker panel;
    • c) determining whether said treatment for Alzheimer's disease is successful by comparing said concentration or amount of each of the biomarkers of the panel in said sample with reference concentrations or amounts of said biomarkers,


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof;

    • and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





Preferably, the neurocognitive disorder in these two aspects of the invention is characterized by tau-toxicity and more preferably the neurocognitive disorder is a tauopathy selected from the group of Alzheimer's disease, frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy (PSP), Pick's disease, corticobasal degeneration, multisystem atrophy (MSA), neurobasal degeneration with iron accumulation, type 1 (Hallervorden-Spatz), argyrophilic grain dementia, Down's syndrome, diffuse neurofibrillary tangles with calcification, dementia pugilistica, Gerstmann-Straussler-Scheinker disease, myotonic dystrophy, Niemann-Pick disease type C, progressive subcortical gliosis, prion protein cerebral amyloid angiopathy, tangle only dementia, postencephalitic parkinsonism, subacute sclerosing panencephalitis, Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex, non-Guamanian motor neuron disease with neurofibrillary tangles/dementia, chronic traumatic encephalopathy, alpha-synucleinopathies, Parkinson's disease or combinations thereof.


In one preferred embodiment the tauopathy is Alzheimer's disease.


In one embodiment, the drug for treating the neurocognitive disorder is a kinase inhibitor; preferably the kinase inhibitor is selected from a tau kinase inhibitor or a casein kinase inhibitor, more preferably a casein kinase 1 alpha, beta, gamma, delta or epsilon.


Even more preferably, the kinase inhibitor is a casein kinase 1 delta inhibitor. Casein kinase 1 delta inhibitors are described in WO2012080727 and WO2012080729 which are incorporated herein as reference.


Examples of casein kinase 1 delta inhibitors are 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine (PS110); 2-amino-3-[(thiophen-2-yl)carbonyl]indolizine-1-carboxamide; 2-[3-(pyridin-4-yl)-1H-pyrazol-4-yl]-1,3-benzoxazole; 2-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (PS278); 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide 2-amino-3-benzoylindolizine-1-carboxamide; 2-amino-1-[(4-fluorophenyl)carbonyl]-1H-indole-3-carboxamide; combinations thereof; or pharmaceutically acceptable salt or solvate thereof.


The most preferred casein kinase 1 delta inhibitor is selected from 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine (PS110); 2-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (PS278); 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; combinations thereof; or pharmaceutically acceptable salt or solvate thereof.


Preferably, step d) in the method of treatment according to the invention further comprises administering an additional therapeutic agent. In one embodiment, the subject has been treated or is being treated with a kinase inhibitor and the additional therapeutic agent is selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof. In another embodiment, the subject has been treated or is being treated with an agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof and the additional therapeutic agent is selected from a kinase inhibitor, preferably a casein kinase 1 delta inhibitor, more preferably a casein kinase inhibitor selected from 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine; 2-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide; combinations thereof; or pharmaceutically acceptable salt or solvate thereof.


In one embodiment of the invention, when assessing the response to a drug for treating a neurocognitive disorder in a subject, wherein the subject has been treated or is being treated with said drug:

    • i) said subject has also been treated or is also being treated with an additional therapeutic agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof; and/or
    • ii) after step c), the method comprises administering an additional therapeutic agent selected from the group of memantine (e.g. Namenda®), galantamine (e.g. Razadyne®), rivastigmine (e.g. Exelon®), donepezil (e.g. Aricept®), solanezumab, 5HT5 antagonists or combinations thereof.


The assaying step a) and/or the measuring step b) of all the methods according to the present invention may further comprise:

    • i) contacting said sample with one or more binding agents to each of said biomarkers of the panel; or
    • ii) detecting in said sample autoantibodies specific to each of said biomarkers; or
    • iii) detecting in said sample by mass spectrometry each of said biomarkers of the panel, optionally by previously labelling said sample with one or more isobaric reactive mass labels; or
    • iv) detecting in said sample by 2D gel electrophoresis each of said biomarkers of the panel; or
    • iv) any combinations of i), ii), iii) or iv).


Preferably, the assaying in step a) and/or the measuring in step b) comprise:

    • i) detecting one or more fragments of said biomarkers in the panel and/or
    • ii) detecting one or more phosphorylated amino acids on tau comprising or having the amino acid sequence of SEQ ID NO: 29 or one or more fragments thereof; wherein when the phosphorylated amino acid on tau to be detected is T181, at least one more phosphorylated amino acid on tau or one or more fragments thereof is detected.


Optionally, the sample is immobilised on a solid support.


The sample to be assayed in the methods according to the present invention is selected from the group of cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue (e.g. brain tissue) or combinations thereof.


Preferably the sample is CSF or blood.


The subject to be diagnosed or assessed or treated may be an animal model (e.g. a rodent or a primate) of AD or of a tauopathy as described herein or a human subject. Preferably, the subject to be diagnosed, assessed or treated is a human subject.


2. Kits

The present invention also provides for kits comprising reagents for assaying and/or measuring in a sample the biomarkers of the panels according to the present invention.


Preferably, the kit allows the diagnosing, staging and assessment of response to a treatment for neurocognitive disorders, in particular Alzheimer's disease.


The reagents of the kits according to the invention may comprise one or more binding agents which specifically bind to the biomarkers of the panels described herein. Preferably, the one or more binding agents are primary antibodies, wherein each primary antibody specifically binds to:

    • i) a different protein of the panel and/or
    • ii) one or more phosphorylated amino acids of tau comprising or having amino acid sequence of SEQ ID NO: 29 or fragments thereof.


More preferably, the primary antibodies are one or more antibodies against protein phosphatase 1 regulatory subunit 14A and/or one or more antibodies against 2′,3′-cyclic-nucleotide 3′-phosphodiesterase. Other primary antibodies include antibodies against the other biomarkers of Groups A, B, C or D and of the proteins listed in Tables 5 to 13.


The primary antibodies may be immobilised on an assay plate, beads, microspheres or particles. Optionally, beads, microspheres or particles may be dyed, tagged or labelled. Optionally the assay plate is a planar array or microtitre multi-well plate.


When the kits comprise primary antibodies against the biomarkers of the panel, the kits may further comprise one or more secondary antibodies which specifically bind to said primary antibodies.


Optionally, the secondary antibodies may be labelled for example fluorescent labelled or tagged.


The kits according to the invention may further comprise one or more detection reagents for detecting the presence of the tagged secondary antibodies.


The sample is preferably selected from the group of cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue (e.g. brain tissue) or combinations thereof.


The kits of the invention allow to:

    • a) assay a sample obtained from a subject for biomarkers of a panel;
    • b) measure in said sample a concentration or an amount of each of the biomarkers of said panel;
    • c) determine whether that subject has a neurocognitive disorder, in particular Alzheimer's disease, by comparing said concentration or amount of each of the protein in said sample to reference concentrations or amounts of said proteins;


      wherein the panel of biomarkers is selected from a panel comprising:


I)





    • i) protein phosphatase 1 regulatory subunit 14A comprising or having the amino acid sequence of SEQ ID NO:1 or an isoform or a variant or a fragment thereof; and/or

    • ii) 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising or having the amino acid sequence of SEQ ID NO:2 or an isoform a variant or a fragment thereof; or


      II) one or more biomarkers selected from Groups A, B, C or D; or


      III) tau or one or more fragments thereof, wherein tau:

    • i) comprises or has the amino acid sequence of SEQ ID NO:29 and

    • ii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422; wherein when the phosphorylated amino acid on tau is T181, the panel comprises tau or one or more fragments thereof having at least one more phosphorylated amino acid; or


      IV) one or more, optionally two or more proteins selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof; or


      V) combinations of I), II), III) and IV.





In particular, the kits according to the invention may instruct to assay (as in step a)) and/or to measure (as in step b)) the sample by:

    • i) contacting said sample with one or more binding agents to each of said biomarkers of the panel; or
    • ii) detecting in said sample autoantibodies specific to each of said biomarkers; or
    • iii) detecting in said sample by mass spectrometry each of said biomarkers of the panel, optionally by previously labelling said sample with one or more isobaric reactive mass labels; or
    • iv) detecting in said sample by 2D gel electrophoresis each of said proteins of the panel; or
    • iv) any combinations of i), ii), iii) or iv).


In yet another embodiment, the kits may comprise reagents suitable for preparing brain tissue, optionally for preparing formalin-fixed paraffin-embedded brain tissue sections.


The kit may additionally provide a reference which provides a quantitative measure by which determination of a concentration or amount of one or more biomarkers can be compared. The reference may indicate the amount or concentration of proteins which indicate the presence or staging or likelihood of developing a neurocognitive disorder such as a tauopathy in particular AD.


The kit may also comprise printed instructions for performing the methods according to the present invention.


In one embodiment, the kit may be for performance of a mass spectrometry assay and may comprise a set of reference peptides (e.g. SRM peptides) in an assay compatible format wherein each peptide in the set is uniquely representative of i) one or more of the biomarkers of Groups A, B, C or D; ii) phosphorylated tau comprising or having the amino acid sequence of SEQ ID NO: 29 or one or more fragments thereof; or iii) one or more of the proteins listed in Tables 5 to 13.


Preferably two or more of such unique peptides are used for each biomarker for which the kit is designed, and wherein each set of unique peptides are provided in known amounts which reflect the amount or concentration of such biomarker in a sample of a healthy subject.


Optionally, the kit may also provide protocols and reagents for the isolation and extraction of the biomarkers according to the invention from a sample, a purified preparation of a proteolytic enzyme such as trypsin and a detailed protocol of the method including details of the precursor mass and specific transitions to be monitored. The peptides may be synthetic peptides and may comprise one or more heavy isotopes of carbon, nitrogen, oxygen and/or hydrogen.


Optionally, the kits of the present invention may also comprise appropriate cells, vessels, growth media and buffers.


3. Detection and Measurement of Biomarkers

The panel of biomarkers described herein comprise both biomarkers where expression is modulated, i.e. quantitatively increased or decreased, and biomarkers which are exclusively present or absent, i.e. qualitatively expressed, in normal versus disease states. The degree to which expression differs in normal versus disease states need only be large enough to be visualised via standard characterisation techniques.


Methods for the detection and quantification of proteins are well known in the art and any suitable method may be employed.


In one embodiment, the biomarkers of the panel may be detected using a binding agent, such as an antibody, specific to that biomarker, for example in an ELISA assay or Western blotting.


Methods relating to the production of antibodies capable of specifically recognising one or more epitopes, including phosphorylated amino acids or amino acids carrying other post-translational modifications, of the individual biomarker in the panel described herein are known in the art. Such antibodies may include, but are not limited to, polyclonal antibodies, monoclonal antibodies (mAbs), humanised or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′)2 fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.


For the production of antibodies, various host animals may be immunised by injection with a protein, or a portion thereof. Such host animals may include, but are not limited to, rabbits, mice and rats. Various adjuvants may be used to increase the immunological response, depending on the host species, including active substances such as lysolecithin, Pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyamin, dinitrophenol, and potentially useful human adjuvant such as BCG bacille Calmette-Fuerin) and Corynebacterium parvum.


Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of animals immunised with an antigen, such as target proteins, or an antigenic functional derivative thereof. For the production of polyclonal antibodies, host animals such as those described above, may be immunised by injection with differentially expressed or pathway protein supplemented with adjuvants as also described above. Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, may be obtained by any technique, which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler and Milstein (1975, Nature 256; 495-497; and U.S. Pat. No. 4,376,110), the human β-cell hybridoma technique (Kosbor, et al., 1983, Immunology Today 4: 72; Cole, et al., 1983, Proc. Natl. Acad. Sci. USA 80; 2026-2030), and the EBV-hybridoma technique (Cole, et al., 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss Inc., pp. 77-96). Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo. Production of high titers of mAbs in vivo makes this the presently preferred method of production.


In addition, techniques developed for the production of ‘chimeric antibodies’ (Morrison, et al., 1984, Proc. Natl. Acad. Sci. 81: 6851-6855; Neuberger, et al., 1984, Nature 312: 604-608; Takeda, et al., 1985, Nature 314: 452-454) by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.


Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; Bird, 1988, Science 242: 423-426; Huston, et al., 1988, Proc. Natl. Acad. Sci. USA 85: 5879-5883; and Ward, et al., 1989, Nature 334: 544-546) can be adapted to produce differentially expressed or pathway protein-single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.


Antibody fragments, which recognise specific epitopes, may be generated by known techniques. For example, such fragments include, but are not limited to, the F(ab′)2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab′)2 fragments. Alternative, Fab expression libraries may be constructed (Huse, et al., 1989, Science 246: 1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.


In some embodiments of the methods described herein, the sample may be immobilised on a solid support for analysis. An antibody sandwich technique may be employed in which binding agents, such as antibodies, specific for the individual protein of the panel are immobilized on a solid support such as a planar surface or a microparticle bead and proteins of the panel are captured by the immobilised binding agents, such as immobilized antibodies. The captured proteins are then detected using a second binding agent, such as a secondary antibody, that may be directly labelled with a signal generating agent (enzyme, fluorescent tag, radiolabel etc.) or may be detected using further amplification (labelled secondary antibody, streptavidin/biotin systems with enzyme, fluorophore, radiolabel etc.). Other methods may include, but are not limited to, one-dimensional or two-dimensional (2D) gel electrophoresis of samples. Such methods are followed by transfer to a solid surface using techniques such as Western blotting and subsequent detection using antibodies specific for the proteins of the panel.


In other embodiments, autoantibodies to the biomarkers of the panel may be detected using the Western blotting approach described above using samples from a healthy subject, a patient or representative of AD, and then detecting the presence of auto-antibodies specific for the biomarkers that are present in the sample, but not in healthy subjects.


An example of a non-antibody binding agent is an aptamer. Examples of aptamers include nucleic acid aptamers and peptide aptamers.


Alternatively, the biomarkers of the panel may be detected by, amongst others, silver staining of 2D gel electrophoresis or mass spectrometry techniques including LS/MS/MS, MALDI-TOF, SELDI-TOF and TMT-SRM.


Other such standard characterisation techniques by which expression differences may be visualised are well known to those skilled in the art. These include successive chromatographic separations of fractions and comparisons of the peaks, capillary electrophoresis, separations using micro-channel networks, including on a micro-chip, SELDI analysis and qPST analysis.


Chromatographic separations can be carried out by high performance liquid chromatography as described in literature, the chromatogram being obtained in the form of a plot of absorbance of light at 280 nm against time of separation. The material giving incompletely resolved peaks is then re-chromatographed and so on.


Capillary electrophoresis may also be employed. The technique depends on applying an electric potential across the sample contained in a small capillary tube. The tube has a charged surface, such as negatively charged silicate glass. Oppositely charged ions (in this instance, positive ions) are attracted to the surface and then migrate to the appropriate electrode of the same polarity as the surface (in this instance, the cathode). In this electro-osmotic flow (EOF) of the sample, the positive ions move fastest, followed by uncharged material and negatively charged ions. Thus, proteins are separated essentially according to charge on them.


Micro-channel networks function similarly to capillaries and can be formed by photoablation of a polymeric material. In this technique, a UV laser is used to generate high energy light pulses that are fired in bursts onto polymers having suitable UV absorption characteristics, for example polyethylene terephthalate or polycarbonate. The incident photons break chemical bonds with a confined space, leading to a rise in internal pressure, mini-explosions and ejection of the ablated material, leaving behind voids which form micro-channels. The micro-channel material achieves a separation based on EOF, as for capillary electrophoresis. It is adaptable to micro-chip form, each chip having its own sample injector, separation column and electrochemical detector.


Surface enhanced laser desorption ionisation time of flight mass spectrometry (SELDI-TOF-MS) combined with ProteinChip technology can also provide a rapid and sensitive means of profiling biomarkers and is used as an alternative to 2D gel electrophoresis in a complementary fashion. The ProteinChip system consists of aluminium chips to which protein samples can be selectively bound on the surface chemistry of the chip (eg. anionic, cationic, hydrophobic, hydrophilic etc). Bound biomarkers are then co-crystallised with a molar excess of small energy-absorbing molecules. The chip is then analysed by short intense pulses of N2 320 nm UV laser with protein separation and detection being by time of flight mass spectrometry. Spectral profiles of each group within an experiment are compared and any peaks of interest can be further analysed using techniques as described below to establish the identity of the protein of the panel.


Isotopic or isobaric Tandem Mass Tags® (TMT® Thermo Scientific, Rockford, USA) technology may also be used to detect proteins of the panel described herein. Briefly, the proteins in the samples for comparison are optionally digested, labelled with a stable isotope tag and quantified by mass spectrometry. In this way, expression of equivalent proteins in the different samples can be compared directly by comparing the intensities of their respective isotopic peaks or of reporter ions released from the TMT® reagents during fragmentation in a tandem mass spectrometry experiment.


Detection of the proteins of the panel described herein may be preceded by a depletion step to remove the most abundant proteins from the sample. The large majority of the protein composition of serum/plasma consists of just a few proteins. For example, albumin, which is present at a concentration of 35-50 mg/ml, represents approximately 54% of the total protein content with IgG adding other 16%. In contrast, proteins changing in response to disease, for example as a result of tissue leakage, may circulate at 10 ng/ml. This vast dynamic range of protein concentrations represents a major analytical challenge and to overcome the problem, a multiple affinity depletion column may be used to remove the most highly abundant proteins (e.g. the 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more highly abundant proteins). This enables the detection of changes in lower abundance ranges because more starting material can be used and there is less interference from the highly abundant molecules. Such a depletion strategy can be applied before any detection method.


4. Examples

Certain aspects and embodiments of the invention will now be illustrated by way of example and with reference to the figures and tables described above. All documents mentioned in this specification are incorporated herein by reference in their entirety for all purposes.


All reagents for sample preparation were purchased from Sigma Aldrich® (Dorset, UK) unless stated. Tandem Mass Tags® (Thermo Scientific®); Acetonitrile (Fisher Scientific®, Loughborough, UK); Trypsin (Roche Diagnostics®, West Sussex, UK).


4.1 Proteins which Correlate with Neurofibrillary Tangle Pathology in the Brain (Braak Staging)


Samples Preparation

Nine frozen tissue samples from inferior temporal cortex samples were dissected from larger frozen tissue sections. Samples were selected according to neurofibrillary tangle (NFT) pathology (=Braak staging) and thus represent all the phases of the AD-related NFT pathology.


The study group comprised one sample each for Braak stage 0, I and II, one sample for Braak stage III, two samples with Braak stage IV, one sample for Braak stage V and a further two samples with Braak stage 6.


The samples were processed and analysed according to the SysQuant® technology as described in details herein below, within one SysQuant® TMT 10plex experiment, which included all nine samples and one study reference.


Quantitative considerations have been performed following a statistical evaluation by a principal component analysis (PCA) of the data, which showed most significant differences in samples with higher Braak staging. Consequently to the PCA results, regulations have been calculated and statistically evaluated by comparing samples with Braak stages V/VI (severe group) to samples with Braak stages III/IV (moderate group).


The cortex samples in lysis buffer (8 M urea, 75 mM NaCl, 50 mM Tris, pH 8.2, protease and phosphatase inhibitors cocktail (Roche) were lysed by sonification (20% Amplitude for 20×1 second, pulsing on and off, on ice (4° C.)) and then centrifuged at 12,500 g for 10 min at 4° C. to eliminate tissue debris. Supernatant was transferred into new tubes and the protein concentration of the samples determined using the Bradford assay. Per sample, the same protein amount was used for all subsequent steps.


After lysis, a reference sample was generated by mixing of identical amounts of all nine individual samples. In particular, for each sample 2.5 mg of protein material were taken and adjusted to a concentration of 3 mg/ml. From each adjusted sample, 167 μL were taken and combined to generate a reference sample. The remaining 666 μL (=2 mg) were used for individual sample manipulation.


DTT was added to each sample (final concentration 5 mM) and incubate for 25 min at 56° C. under shaking to reduce disulfide bonds. Samples were then allowed to cool at room temperature before adding iodoacetamide (final concentration 14 mM) and incubated for 30 min at room temperature and in the dark to alkylate cysteine residues. Unreacted iodoacetamide was quenched by adding DTT to additional 5 mM and incubating 15 min at room temperature in the dark.


Samples were diluted with 25 mM Tris-HCl, pH 8.2, to reduce the concentration of urea to 1.6 M. Trypsin (Roche, UK) was added to a final minimum concentration of 5 ng/μL (trypsin to substrate ratio of at least 1:100) together with CaCl2 (final concentration of 1 mM). Samples were incubated at 37° C. overnight with shaking (˜15-18 hours). Digested samples were allowed to cool to room temperature and digestion was stopped by acidification with TFA to 0.4% (vol/vol). Samples were centrifuged at 2,500 g for 10 min at room temperature and the pellet was discarded.


Samples were desalted using a 200 mg SepPak tC18 cartridges (Thermo Scientific UK) according to manufacturer's instructions and proteins were eluted with elution buffer (50% ACN, 50% H2O) and concentrated to dryness in Speedvac.


For TMT® labelling samples were re-solubilised into 567 μL of TEAB/ACN buffer for a final concentration of 2.0-2.5 mg total peptide amount per sample. TMT® labels from the TMT® 10plex regent set was added to each sample according to Table 14 below to give a final concentration of 15 mM TMT® in each sample.











TABLE 14





Sample No.
Braak staging
TMT ® labelling







1
V
TMT6-126


2
IV
TMT6-128


3
VI
TMT6-129e


4
I
TMT6-127


5
VI
TMT6-130


6
0
TMT6-128e


7
II
TMT6-129


8
IV
TMT6-127e


9
III
TMT6-130e


Ref.
N/A
TMT6-131





Ref. = Reference sample containing an aliquot of all the samples






Reaction were allowed to take place for 1 hr at room temperature. Hydroxylamine was added to each sample to a final concentration of 0.25% [w/v] hydroxylamine and incubated for 15 minutes. Samples were then diluted 1:3 with 2% TFA and then further diluted with water to reduce the concentration of ACN to below 5%. Samples were mixed in equal amounts to achieve one SysQuant10plex sample which was then split into two aliquots which were desalted (500 mg SepPak tC18 cartridges) and fractionated by SCX chromatography (3 mL/min as flow rate; Buffer A: Water+0.1% TFA; Buffer C: 7 mM KH2PO4, pH 2.65, 30% ACN (vol/vol); Buffer D: 7 mM KH2PO4, 350 mM KCl, pH 2.65, 30% ACN (vol/vol)) according to the protocol in Table 15.














TABLE 15







Time
Buffer A
Buffer C
Buffer D



[min]
[%]
[%]
[%]





















0
0
100
0



2
0
100
0



35
0
75
25



36
0
0
100



46
0
0
100



47
100
0
0



57
100
0
0



58
0
100
0



67
0
100
0



68
0
100
0










Lyophilized peptides from desalting were re-suspended in 800 μL of buffer A, injected onto the HPLC system. Twelve fractions were collected and “smart pooled” by mixing fractions with low numbers of peptides with fractions containing large numbers of peptides to provide 6 final fractions with similar total peptide content and desalted.


Then, small portions were taken from each fraction to apply the subsequent analysis of non-enriched fractions. The remaining portion of the fractions were applied to enrichment for phosphopeptides by either the IMAC or the TiO2 procedures, both well-known in the art. Dried fractions were transferred for LC-MS analysis


LC-MS/MS Analysis

“Smart-pooled” fractions were analysed twice each by LC-MS/MS (double-shot workflow), with MS acquisition performed by Top Speed MS2 HCD method.


Peptides from non-enriched fractions were re-suspended in 100 μl 2% ACN/0.1% FA, then 5 ul per fraction was injected onto a 2 cm×75 um Acclaim PepMap100 pre-column, and separated using an EASY-Spray 50 cm×75 um ID, PepMap RSCL, C18, 2 um, on the EASY-nLC 1000 system (Thermo Fisher Scientific). Peptides were resolved using a 160 min separation gradient of 8 to 30% ACN/0.1% FA at 200 nL/min.


Peptides from all phospho-enriched fractions were re-suspended in 30 μl of 2% ACN/0.1% FA, then 5 μL per fraction injected and resolved using a 160 min separation gradient of 10 to 30% 0.1% FA in ACN at 200 nL/min.


Mass spectra were acquired on an Orbitrap Fusion™ Tribrid™ Mass Spectrometer (Thermo Fisher Scientific) for a total run time of 180 min using top speed higher collision induced dissociation (HCD) FTMS2 scans at 30,000 resolving power, following each FTMS scan (120,000 resolving power). HCD was carried out on the most intense ions from each FTMS scan, and then put on a dynamic exclusion list for 30 sec to avoid repeated sequencing of the same analyte. Each sample was analysed by two LC-MS/MS analytical repeats (double shot workflow).


Computational MS

The acquired spectra were processed using Proteome Discoverer 1.4 (PD 1.4; Thermo Fisher Scientific) software using the human specific UniProtKB/Swiss-Prot database (88,647 sequence entries) downloaded on 22 Feb. 2014. The raw data was searched using the Sequest HT and Mascot (Matrix Mascot server 2.2.06) search algorithms within PD 1.4. MS raw data files that belonged to the 6 fractions of any enrichment arm of the SysQuant® workflow and those which belonged to the same analytical set of runs were submitted as an individual MudPit search each. Thus, in total four MudPit searches were performed. After filtering the search results at 1% false discovery rate (FDR) at the peptide level and at least one rank 1 peptide per protein, search results were exported to MS Excel files. This data was processed by defined scripts to list all identified peptides, and their protein origin.


Data Analysis

All protein identification, peptide sequence, phosphorylation site information along with quantitative values and biological information relating to the peptide, protein and phosphorylation sites were assembled in a Microsoft Excel file (QuantSheet) to allow structured data analysis based on fold-change, significance, biological function and cellular localisation.


To evaluate the general data quality by bioinformatic means, i.e. to visualize the effect of Braak staging, and thus to guide subsequent quantitative computations, a principle component analysis (PCA) was carried out.


A thorough biological data interpretation was also performed and a targeted analysis of proteins of interest was applied, for example for tau protein and additional AD biomarkers Proteins implicated in neurodegeneration.


4.2 Quantification of Regulated Proteins in AD CSF Using TMTcalibrator™

We also identified the presence and relative abundance of phosphorylated peptides of tau in human CSF drawn from controls that were biochemically negative for AD and those that were biochemically positive. Briefly, post-mortem collected pre-frontal cortex material from human subjects with moderate tau pathology (Braak stages III-IV; n=3) or severe tau pathology (Braak stages V-VI; n=3), performed in separate experiments, were pooled, digested with trypsin and labelled in four separate aliquots with TMT reagents TMT10-129C, 130N, 130C and 131. Each labelled aliquot from the same brain digest pool was mixed at a ratio of 0.3 mg:1.2 mg:1.8 mg:3.0 mg respectively to form a calibration standard. At the same time human cerebrospinal fluid samples (600 ug protein content (600 ul) per individual) from three non-cognitively impaired control individuals and three biochemically diagnosed cases of AD were digested with trypsin and labelled with TMT10 reagents TMT10-126, 127N, 127C, 128N, 128C and 129N respectively and mixed to form the clinical test sample. Finally an equal volume of the calibration standard and clinical test sample were mixed to form the analytical sample according to FIG. 7.


Mass spectrometry and data analysis was carried out as described above in Section 4.1.


4.3 Effects of Tau Inhibition In Vivo

The inventors have also applied SysQuant to analyse protein and phosphorylation changes in the brains of TMHT (Thy-1 Mutated Human Tau) mice (developed by QPS® Austria, http://www.qps-austria.com) treated with small molecule inhibitors of the tau kinase Casein Kinase 1 delta (CK1d). Starting at 8.5 months (±2 weeks) of age, TMHT mice received CK1d inhibitors 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (PS278-05), 5-(1,3-benzoxazol-2-yl)-4-(pyridine-4-yl)pyrimidin-2-amine (PS110), compound PF4800567 (3-[(3-Chlorophenoxy)methyl]-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine; Tocris®) or vehicle (0.5% w/v methylcellulose) for 8 weeks (54 applications) at a dosage of 30 mg/kg body weight, orally via gavage.


In total 48 animals were used and allocated to 4 treatment groups. Table 16 describes the animals, cohort and treatment group allocation, sex and age of the animals.


Brain samples were collected within 10 minutes of euthanasia, washed in ice cold saline, transferred to a clean Eppendorf tube and immediately frozen in liquid nitrogen.


One hippocampus sample (left or right) from each of three animals in each of the vehicle, PS110 and PS278-05 groups were selected for analysis by SysQuant. A pool of all nine samples was also prepared as a reference channel. All TMT® labelling, mixing and mass spectrometry and bioinformatics studies were performed essentially as described in Section 4.1. For peptide and protein identification the UniProtKB mouse database was used, supplemented by inclusion of all human tau isoform entries.


All protein identification, peptide sequence, phosphorylation site information along with quantitative values and biological information relating to the peptide, protein and phosphorylation sites were assembled in a Microsoft Excel file (QuantSheet) to allow structured data analysis based on fold-change, significance, biological function and cellular localisation.


4.4 Detection of Phosphorylated Tau

The kinetic profile of tau phosphorylation in the various human and mouse samples studied in Sections 4.1, 4.2 and 4.3 was determined by a targeted data analysis of the respective QuantSheets. All peptides that matched the human MAPT (P10636; including P10636-8=SEQ ID NO:29) UniProt sequences were exported into a separate Microsoft Excel sheets.


Ion intensity values for each non-phosphorylated peptide were summed and the average calculated for each disease severity group (Braak Stage 0-II; Braak Stage III-IV; Braak Stage V-VI) to give a value for total tau expression. For each phosphorylated amino acid e.g. pT181, the ion intensity values of all peptides containing said phosphorylated amino acid were summed and the average calculated for each disease severity group (Braak Stage 0-II; Braak Stage III-IV; Braak Stage V-VI).


A total of 185 unique peptides from human tau were quantified in all nine brain samples with 35 high confidence (phosphoRS score >75%) phosphorylation sites. Of these, the levels of 74 peptides were significantly (p<0.05) regulated in the brains of patients with moderate (Braak stage III-IV) and severe (Braak stage V-VI) tau pathology


Similarly, the expression levels of total tau and for each phosphorylated serine, threonine and tyrosine amino acid in mouse brains following treatment with vehicle control or the CK1d inhibitors PS278-05 and PS110 were calculated.


All peptides that matched the human MAPT (P10636; SEQ ID NO:29) UniProt sequence and/or the mouse MAPT (P10637) were exported into a separate Microsoft Excel sheet. Ion intensity values for each phosphopeptide were summed and the average calculated for each treatment group (Vehicle control, PS110, PS278-05). We quantified 124 unique peptides from human tau in all nine brain samples with 39 high confidence (phosphoRS score >75%) phosphorylation sites. Of these, the levels of 37 peptides were significantly (p<0.05) regulated in the hippocampi of mice treated with PS110 whilst 22 peptides were significantly (p<0.05) regulated in the hippocampi of mice treated with PS278-05.


Finally, the levels of total tau and for each phosphorylated serine, threonine and tyrosine amino acid in human CSF samples drawn from control and biochemically confirmed cases of AD were determined as above. All peptides that matched the human MAPT (P10636) UniProt sequence were exported into a separate Microsoft Excel sheet. Ion intensity values for each phosphopeptide were summed and the average calculated for each group (Control, AD). There were 65 quantified tau peptides in human CSF covering 27 high confidence phosphorylation sites.


In total, across the three studies we identified 44 unique phosphorylation sites on tau protein with 19 sites being quantified in all samples of human brain, mouse brain and human CSF. Table 4 collates all Tau phosphorylation sites identified by SysQuant® and TMTcalibrator™ in mouse and human brain tissue and human CSF. Amino acid numbering based on human 2N4R tau (Uniprot Accession Number P10636-8 with SEQ ID NO:29).


The method of measurement of these phosphorylated residues in tau protein are not intended to limit the invention. One or more of the sites may be measured using a binding agent such as an antibody or aptamer specific for the phosphorylated residue. However, for some sites the use of a binding agent will be less desirable due to the influence of adjacent phosphorylation on additional residues. One such case is the measurement of phosphorylated threonine 181 (2N4R tau numbering). Current diagnosis of AD is supported by the measurement of CSF levels of total tau and pT181 to yield a ratio. The larger the ratio the greater the probability that the patient has AD. In the present study we have identified that pT181 can be measured on two separate peptides in CSF. One is a singly phosphorylated peptide at threonine 181 whilst the other is triply phosphorylated at threonine 181, serine 184 and serine 185. It is probable that additional phosphorylation at 5184 and 5185 will interfere with the affinity of the binding agent reducing its ability to bind to the triply phosphorylated species and so under-representing the total amount of pT181 in CSF.


To overcome these limitations, phosphorylated tau is preferably measured using mass spectrometry. Any form of mass spectrometry capable of providing a relative or absolute quantification of each phosphorylation may be used. Such methods include but are not limited to data independent acquisition (DIA), data dependent acquisition (DDA), selected reaction monitoring (SRM), multiple reaction monitoring (MRM), or TMTcalibrator™. In each case a reference phosphopeptide that can be differentiated from the endogenous phosphotau peptide in the test sample using mass spectrometry is provided. The reference phosphopeptide may be provided from a biological sample and made distinct from endogenous phosphopeptides using an isotopic or isobaric mass tag. Alternatively the reference phosphopeptide may be generated from digestion of a recombinant tau protein manufactured using a synthetic lysine source that has several H2, C13, N15, O18 atomic substitutions to ensure each tryptic peptide will have a mass greater than the naturally occurring equivalent phosphopeptide of at least 1 Dalton, preferably more than 2 Daltons and most preferably more than 5 Daltons. Fully synthetic peptides manufactured by sequential amino acid addition may be used wherein one or more amino acids within the peptide sequence contains atomic substitutions with H2, C13, N15, O18 or other such appropriate stable heavy isotope.


Irrespective of the means of detection and quantification, assays intended for early diagnosis, prognosis of disease progression or monitoring of therapeutic effect may measure one, two or more different tau phosphopeptides.


4.5 Identification of Other Brain-Derived Proteins in Human CSF

In addition to identifying regulated peptides derived from tau protein, we also analysed the data from Example 4.2 to identify other disease-related proteins that are differentially expressed in human AD CSF. Briefly, the TMT® reporter ion intensities for all unmodified peptides matched to a unique UniProt accession number were summed and used to determine the relative expression of the relevant protein in human CSF. Based on the summed intensity values a log 2 ratio was calculated for each protein expression in control (n=3) and AD (n=3) groups. Statistical significance was calculated as a p-value based on the six independent protein quantification values using a two sample t-test. The resulting data matrix was exported to Microsoft Excel and filtered to select all proteins showing greater than 40% regulation (−0.5≤log 2≥0.5) and p≤0.05. This was performed for both TMTcalibrator™ experiments using brain calibrants with moderate and severe tau pathology. The list of regulated proteins including log 2 ratio and p-values is shown in Table 5.


Any of the proteins listed in Table 5 may be used as diagnostic and/or prognostic biomarkers of AD.


4.6 Results

The present inventors have surprisingly identified biomarkers which are highly regulated in the brain of patients with AD and/or which are present in the CSF and/or which are highly regulated in response to administration of casein kinase inhibitors. In addition, tau toxicity in the brain of the patients used in these experiments present elevated expression and hyperphosphorylation (tau toxicity).


Surprisingly, when the data were analysed with respect to Braak stages V or VI, protein phosphatase 1 regulatory subunit 14A resulted to be the most upregulated protein (FIG. 3). Interestingly, levels of this protein first fell between mild (Braak stage 0-II) and moderate (Braak stage III/IV) before rising significantly in severe disease. Treatment with both casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridine-4-yl)pyrimidin-2-amine or 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide resulted in an up-regulation of protein phosphatase 1 regulatory subunit 14A, apparently counteracting the drop in expression levels seen in moderate tau pathology in humans). FIG. 3 shows a Venn diagram for the analyses performed on the brains of patients with AD (label “Human Brain”), CSF of patients with AD at Braak stage V or VI (label “Human CSF 5/6) and on the brains of mice treated with 5-(1,3-benzoxazol-2-yl)-4-(pyridine-4-yl)pyrimidin-2-amine (upper diagram) or 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (lower diagram) (label “Mice ck1 inh.). The numbers in the brackets below each of the labels indicate the number of proteins identified. As it can be seen in FIG. 3, there is only one protein which is upregulated in all the samples and this protein is protein phosphatase 1 regulatory subunit 14A.


Even more surprisingly, when the data were analysed with respect to Braak stages V or VI, 2′,3′-cyclic-nucleotide 3′-phosphodiesterase resulted to be the most upregulated protein (FIG. 4). Interestingly, levels of this protein first fell between mild (Braak stage 0-II) and moderate (Braak stage III/IV) before rising significantly in severe disease. Again, in the parallel study in the TMHT model of tauopathy the type of treatment did not play a significant role with both compounds increasing the expression of the protein. FIG. 4 shows a Venn diagram for the analyses performed on the brains of patients with AD (label “Human Brain”), CSF of patients with AD at Braak stage 3 or 4 (label “Human CSF 3/4) and on the brains of mice treated with 5-(1,3-benzoxazol-2-yl)-4-(pyridine-4-yl)pyrimidin-2-amine (upper diagram) or 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide (lower diagram) (label “Mice ck1 inh.). As it can be seen in FIG. 4, there is only one protein which is upregulated in all the samples and this protein is 2′,3′-cyclic-nucleotide 3′-phosphodiesterase.



FIG. 5 shows a bar diagram (A) which depicts the levels of protein phosphatase 1 regulatory subunit 14A levels in human brain with mild (Braak 0-II) (n=3), moderate (Braak III/IV) or severe (Braak V/VI) tau pathology; the levels of protein phosphatase 1 regulatory subunit 14A are increased in the brain of human subjects with an advanced stage of the neurocognitive disorder (Braak V/VI). FIG. 5 diagram (B) shows that mouse brain from the TMHT model of human tauopathy treated orally with vehicle alone or vehicle including 30 mg/kg of casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine and 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide responded to treatment altering the concentration or amount of protein phosphatase 1 regulatory subunit 14A. Finally, FIG. 5 diagram (C) shows that CSF from patients with biochemically diagnosed AD (n=3) have a decreased level of protein phosphatase 1 regulatory subunit 14A compared to cognitively affected non-AD controls (n=3).



FIG. 6 shows a bar diagram (A) which depicts the levels of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase levels in human brain with mild (Braak 0-II) (n=3), moderate (Braak III/IV) or severe (Braak V/VI) tau pathology; the levels of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase are increased in the brain of human subjects with an advanced stage of the neurocognitive disorder (Braak V/VI). FIG. 6 diagram (B) shows that mouse brain from the TMHT model of human tauopathy treated orally with vehicle alone or vehicle including 30 mg/kg of casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine and 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide responded to treatment altering the concentration or amount of protein 2′,3′-cyclic-nucleotide 3′-phosphodiesterase. Finally, FIG. 6 diagram (C) shows that CSF from patients with biochemically diagnosed AD (n=3) have a increased level of 2′,3′-cyclic-nucleotide 3′-phosphodiesterase compared to cognitively affected non-AD controls (n=3).


In summary, 1) both phosphatase 1 regulatory subunit 14A and 2′,3′-cyclic-nucleotide 3′-phosphodiesterase are elevated in the brain of subjects with an advanced stage of a neurocognitive disorder (Braak stage V or stage VI); 2) in the CSF of AD subjects protein phosphatase 1 regulatory subunit 14A is decreased whilst 2′,3′-cyclic-nucleotide 3′-phosphodiesterase is elevated in comparison to cognitively affected non-AD controls; and 3) both phosphatase 1 regulatory subunit 14A and 2′,3′-cyclic-nucleotide 3′-phosphodiesterase are upregulated in response to casein kinase 1 delta inhibitors 5-(1,3-benzoxazol-2-yl)-4-(pyridin-4-yl)pyrimidin-2-amine and 2-Methyl-amino-3-[(4-fluorophenyl)carbonyl]indolizine-1-carboxamide.


Finally, additional proteins from Groups A, B, C, D and tables 5 to 13 can also be selected together with phosphatase 1 regulatory subunit 14A and 2′,3′-cyclic-nucleotide 3′-phosphodiesterase to form enlarged biomarker panels for the diagnoses, staging and assessment of treatment of a neurocognitive disorder, in particular of AD.

Claims
  • 1-30. (canceled)
  • 31. A kit comprising reagents for assaying and/or measuring in a sample one or more biomarkers of a panel comprising: i) a protein phosphatase 1 regulatory subunit 14A comprising the amino acid sequence of SEQ ID NO:1 or an isoform, a variant or a fragment thereof; and/orii) a 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising the amino acid sequence of SEQ ID NO:2 or an isoform, a variant or a fragment thereof.
  • 32-36. (canceled)
  • 37. The kit according to claim 31, wherein the reagents comprise a set of reference peptides in an assay compatible format and each reference peptide is uniquely representative of a single biomarker in the panel.
  • 38. The kit according to claim 37, wherein the set of reference peptides comprises at least two uniquely representative peptides for each biomarker in the panel, and wherein the uniquely representative peptides are provided in known amounts which reflect the amount or concentration of each biomarker in a sample of a healthy subject.
  • 39. The kit according to claim 37, wherein the reference peptides are synthetic peptides.
  • 40. The kit according to claim 37, wherein the reference peptides comprise one or more heavy isotopes of carbon, nitrogen, oxygen and/or hydrogen.
  • 41. The kit according to claim 31, wherein the panel further comprises one or more biomarkers selected from Group A, which comprises Actin alpha cardiac muscle 1 comprising the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising the amino acid sequence of SEQ ID NO:3, cAMP-dependent protein kinase type I-beta regulatory subunit comprising the amino acid sequence of SEQ ID NO:24, Catenin delta-1 comprising the amino acid sequence of SEQ ID NO:4, Centrosomal protein of 170 kDa comprising the amino acid sequence of SEQ ID NO:23, Clathrin light chain B comprising the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising the amino acid sequence of SEQ ID NO:14, GMP reductase 1 comprising the amino acid sequence of SEQ ID NO:27, Guanine nucleotide-binding protein G(q) subunit alpha comprising the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising the amino acid sequence of SEQ ID NO:15, KxDL motif-containing protein 1 comprising the amino acid sequence of SEQ ID NO:28, Lambda-crystallin homolog comprising the amino acid sequence of SEQ ID NO:18, Myelin-associated oligodendrocyte basic protein comprising the amino acid sequence of SEQ ID NO:20, Neutral alpha-glucosidase AB comprising the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising the amino acid sequence of SEQ ID NO:16, Protein KIAA1045 comprising the amino acid sequence of SEQ ID NO:25, Secernin-2 comprising the amino acid sequence of SEQ ID NO:8, Serum albumin comprising the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising the amino acid sequence of SEQ ID NO:9, Synaptoporin comprising the amino acid sequence of SEQ ID NO:22, Syntaphilin comprising the amino acid sequence of SEQ ID NO:10, Transmembrane protein 119 comprising the amino acid sequence of SEQ ID NO: 21 and Tubulin alpha chain-like 3 comprising the amino acid sequence of SEQ ID NO:26.
  • 42. The kit according to claim 41, wherein the biomarker panel further comprises one or more biomarkers selected from Groups B, C or D.
  • 43. The kit according to claim 31, wherein the biomarker panel further comprises one or more of a tau protein or a fragment thereof, wherein the tau protein: i) comprises the amino acid sequence of SEQ ID NO:29 andii) comprises one or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422 of SEQ ID NO:29;wherein when the phosphorylated amino acid is T181, the panel further comprises a tau protein or a fragment thereof having at least one more phosphorylated amino acid.
  • 44. The kit according to claim 31, wherein the biomarker panel further comprises one or more biomarkers selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13, or a combination thereof.
  • 45. The kit according to claim 31, wherein the reagents comprise one or more binding agents, wherein each binding agent binds specifically to one of the biomarkers of the panel.
  • 46. The kit according to claim 31, wherein the sample is selected from cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue, or a combination thereof.
  • 47. A method for measuring in a sample the concentration or amount of one or more biomarkers of a panel, comprising: i) a protein phosphatase 1 regulatory subunit 14A comprising the amino acid sequence of SEQ ID NO:1 or an isoform, a variant or a fragment thereof; and/orii) a 2′,3′-cyclic-nucleotide 3′-phosphodiesterase comprising the amino acid sequence of SEQ ID NO:2 or an isoform, a variant or a fragment thereof;said method comprising at least one of the following steps: a) detecting in the sample by mass spectrometry each of the one or more biomarkers of the panel or fragments thereof;b) contacting the sample with one or more binding agents to each of the biomarkers of the panel; and/orc) detecting in the sample by 2D gel electrophoresis each of the biomarkers of the panel.
  • 48. The method according to claim 47, wherein step a) and/or step b) comprises detecting one or more fragments of the biomarkers in the panel.
  • 49. The method according to claim 47, wherein the sample is immobilized on a solid support.
  • 50. The method according to claim 47, wherein the sample is selected from cerebrospinal fluid (CSF), blood, plasma, serum, saliva, urine, tissue or combinations thereof taken from a human subject.
  • 51. The method according to claim 47, wherein the method is performed using the kit of claim 31.
  • 52. The method according to claim 47, further comprising measuring in the sample the concentration or amount of one or more biomarkers selected from Group A, which comprises Actin alpha cardiac muscle 1 comprising the amino acid sequence of SEQ ID NO:11, Antithrombin-III comprising the amino acid sequence of SEQ ID NO:12, BH3-interacting domain death agonist comprising the amino acid sequence of SEQ ID NO:3, cAMP-dependent protein kinase type I-beta regulatory subunit comprising the amino acid sequence of SEQ ID NO:24, Catenin delta-1 comprising the amino acid sequence of SEQ ID NO:4, Centrosomal protein of 170 kDa comprising the amino acid sequence of SEQ ID NO:23, Clathrin light chain B comprising the amino acid sequence of SEQ ID NO:5, Egl nine homolog 1 comprising the amino acid sequence of SEQ ID NO:13, Fibrinogen gamma chain comprising the amino acid sequence of SEQ ID NO:14, GMP reductase 1 comprising the amino acid sequence of SEQ ID NO:27, Guanine nucleotide-binding protein G(q) subunit alpha comprising the amino acid sequence of SEQ ID NO:6, Insulin-like growth factor-binding protein 6 comprising the amino acid sequence of SEQ ID NO:15, KxDL motif-containing protein 1 comprising the amino acid sequence of SEQ ID NO:28, Lambda-crystallin homolog comprising the amino acid sequence of SEQ ID NO:18, Myelin-associated oligodendrocyte basic protein comprising the amino acid sequence of SEQ ID NO:20, Neutral alpha-glucosidase AB comprising the amino acid sequence of SEQ ID NO:7, Nuclear pore complex protein Nup155 comprising the amino acid sequence of SEQ ID NO:19, OCIA domain-containing protein 1 comprising the amino acid sequence of SEQ ID NO:16, Protein KIAA1045 comprising the amino acid sequence of SEQ ID NO:25, Secernin-2 comprising the amino acid sequence of SEQ ID NO:8, Serum albumin comprising the amino acid sequence of SEQ ID NO:17, Short-chain specific acyl-CoA dehydrogenase comprising the amino acid sequence of SEQ ID NO:9, Synaptoporin comprising the amino acid sequence of SEQ ID NO:22, Syntaphilin comprising the amino acid sequence of SEQ ID NO:10, Transmembrane protein 119 comprising the amino acid sequence of SEQ ID NO: 21 and Tubulin alpha chain-like 3 comprising the amino acid sequence of SEQ ID NO:26.
  • 53. The method according to claim 47, further comprising measuring in the sample the concentration or amount of one or more biomarkers selected from Groups B, C or D.
  • 54. The method according to claim 47, further comprising measuring in the sample the concentration or amount of: a) a tau protein or a fragment thereof, wherein the tau protein: i) comprises the amino acid sequence of SEQ ID NO:29 andii) comprises one or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422 of SEQ ID NO:29;wherein when the phosphorylated amino acid is T181, the panel further comprises a tau protein or a fragment thereof having at least more phosphorylated amino acid; orb) one or more biomarkers selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13, or combinations thereof.
  • 55. A kit comprising reagents for assaying and/or measuring in a sample: a) one or more biomarkers of a panel comprising a tau protein or a fragment thereof, wherein the tau protein: i) comprises the amino acid sequence of SEQ ID NO:29 andii) comprises one or more, optionally two or more phosphorylated amino acids selected from T39, S46, T50, T52, T56, S61, T63, S64, S68, T69, S113, T181, S184, S185, S191, S195, S198, S199, S202, S205, S208, S210, T212, S214, T217, T231, S235, S237, S238, S258, S262, S285, S289, S356, Y394, S396, S400, T403, S404, S409, S412, S413, T414/S416 or S422 of SEQ ID NO:29;wherein when the phosphorylated amino acid is T181, the panel further comprises a tau protein or a fragment thereof having at least one more phosphorylated amino acid; orb) one or more biomarkers selected from Tables 5, 6, 7, 8, 9, 10, 11, 12, 13 or combinations thereof.
Priority Claims (2)
Number Date Country Kind
1509134.1 May 2015 GB national
1512596.6 Jul 2015 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2016/062090 5/27/2016 WO 00