WO2011143574A2 - Plasma biomarkers for diagnosis of alzheimer's disease - Google Patents

Plasma biomarkers for diagnosis of alzheimer's disease Download PDF

Info

Publication number
WO2011143574A2
WO2011143574A2 PCT/US2011/036457 US2011036457W WO2011143574A2 WO 2011143574 A2 WO2011143574 A2 WO 2011143574A2 US 2011036457 W US2011036457 W US 2011036457W WO 2011143574 A2 WO2011143574 A2 WO 2011143574A2
Authority
WO
WIPO (PCT)
Prior art keywords
disease
alzheimer
individual
biomarkers
age
Prior art date
Application number
PCT/US2011/036457
Other languages
French (fr)
Other versions
WO2011143574A3 (en
Inventor
John Q. Trojanowski
Virginia M.Y. Lee
Leslie M. Shaw
Holly D. Soares
Eve H. Pickering
Andrew Kuhn
William Tzu-Lung Hu
Original Assignee
The Trustees Of The University Of Pennsylvania
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Trustees Of The University Of Pennsylvania, Pfizer Inc. filed Critical The Trustees Of The University Of Pennsylvania
Publication of WO2011143574A2 publication Critical patent/WO2011143574A2/en
Publication of WO2011143574A3 publication Critical patent/WO2011143574A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • Alzheimer's disease is a progressive neurodegenerative disorder estimated to affect 27 million people worldwide with numbers doubling every 20 years (Prince et al, 2009 Alzheimer's Disease International 1-92). Although symptoms of Alzheimer's disease manifest early as deficits in memory and other cognitive domains, pathological data show neuropathological features of Alzheimer's disease, including amyloid plaques and neurofibrillary tangles, occur well before the onset of dementia (Sawa et al, 2009 N Engl J Med. 360(22):2302-2309). Research focused upon cognitive performance in individuals who have progressed to dementia have further specified a pre-demented stage characterized by deficits in memory and executive function (Petersen et al, 2008 CNS Spectr. 13(l):45-53).
  • Alzheimer's disease a progressive neurodemented stage of Alzheimer's disease
  • a stereotypical biomarker signature supporting the notion that Alzheimer's disease can be diagnosed prior to the onset of dementia and that clinical studies focused upon dementia prevention are feasible (Craig-Schapiro et al, 2009 Neurobiol Dis. 35(2): 128-140).
  • DSM-IV-TR Diagnostic and Statistical manual of Mental Disorders
  • NINCDS-ADRDA National Institute of Neurological Disorders and Stroke - Alzheimer's Disease and Related Disorders
  • NINCDS-ADRDA are similar, but specify that onset is gradual and other systemic or neural diseases known to impair cognition have been excluded (McKhann et al., 1984 Neurology 34(7): 939-944).
  • Most clinical studies in Alzheimer's disease utilize either NINCDS-ADRDA or DSM-IV criteria to enroll mild to moderate stage individuals.
  • individuals with dementia are already in mid to late neuropathological stages of Alzheimer's disease.
  • Alzheimer's disease-related neuropathology is reversible once dementia manifests. Indeed, recent studies have implied that amelioration of plaque load may occur following therapeutic treatment, but such clearance may not be associated with clinical benefit once the underlying pathology is significant.
  • symptomatic individuals at risk of progressing to dementia do exhibit a stereotypical biomarker phenotype.
  • symptomatic, and in some cases non-symptomatic, individuals at risk of progressing to dementia exhibit decreased levels of CSF amyloid beta peptide 42 ( ⁇ 42) and elevated levels of tau and phosphorylated tau (Mattsson et al, 2009 Jama 302(4):385-393; Shaw et al, 2009 Ann Neurol. 65(4):403-413; Sunderland et al, 2003 Jama 289(16):2094-2103; Visser et al, 2009 Lancet Neurol.
  • CSF and imaging biomarkers hold great promise as tools to enable early Alzheimer's disease drug development, they are problematic as diagnostic tools for the general practitioner.
  • CSF procedures in many countries are considered highly invasive and PET imaging techniques are expensive and, in some countries, not widely available.
  • Simple cognitive tests offer an alternative screening approach, but require time that is often not reimbursed by many health care systems. In the setting of a general practitioner where time constraints with individuals are an issue, cognitive tests are often not brief enough to accommodate a standard six-ten minute individual interview.
  • a blood test would be a simple route for screening, and, if positive, could be used in combination with more extensive cognitive testing followed by referral to a specialist for more confirmatory CSF and imaging testing that would enable early diagnosis.
  • a simple blood test would offer a tractable option towards screening individuals at risk.
  • the present invention fills these needs by providing among other things, novel biomarkers and combinations of biomarkers useful for diagnosing Alzheimer's disease, as well as methods and kits for using the biomarkers to diagnose Alzheimer's disease.
  • the present invention provides a composition for detecting Alzheimer's
  • the biological sample is a plasma sample.
  • the biomarkers for Alzheimer's disease comprise Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
  • the biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309.
  • the biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
  • Hepatocyte Growth Factor IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
  • Thrombomodulin tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
  • TRIP-1 tissue inhibitor of metalloproteinases-1
  • VCAM-1 vascular cell adhesion molecule- 1
  • VEGF Vascular endothelial growth factor
  • Von Willebrand Factor Von Willebrand Factor
  • the biomarkers include one or more biomarkers selected from Table 2.
  • the reagent for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual is an antibody. In one embodiment, the reagent is on a solid support.
  • the invention provides a method of diagnosing Alzheimer's Disease in an individual, the method comprising analyzing a biological sample from an individual to determine the level(s) of one or more biomarkers for Alzheimer's Disease in the sample, wherein the one or more biomarkers are selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to diagnose whether the individual has Alzheimer's Disease.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to diagnose whether the individual has Alzheimer's Disease.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
  • the algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K- Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
  • the Alzheimer's Disease diagnosed is selected from the group consisting of late onset Alzheimer's disease, early onset Alzheimer's disease, familial Alzheimer's disease, and sporadic Alzheimer's disease.
  • the biological sample is selected from the group consisting of whole blood, a blood component, CSF, urine, and any combination thereof.
  • the present invention provides a method for determining whether dementia in an individual is associated with AD, the method comprising analyzing a biological sample from an individual to determine the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin in the sample, and comparing the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP-2, and resistin in a corresponding reference sample to determine whether dementia in an individual is associated with AD.
  • the dementia is associated with AD.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine the presence or status of dementia in the individual.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • the biological sample from the individual is analyzed to determine the level(s) of one or more biomarkers selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to determine the presence or status of Alzheimer's Disease or other types of dementia in the individual.
  • the levels of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, resistin biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine the presence or status of Alzheimer's Disease or other types of dementia in the individual.
  • the present invention also includes a method for diagnosing Alzheimer's disease in an individual, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample from the individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive control reference values and optionally to negative control reference values, wherein levels of the biomarkers in the plasma sample of the individual resemble the positive control reference values and do not resemble the negative control reference values are an indication that the individual has Alzheimer's disease.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the test individual in order to diagnose whether the individual has Alzheimer's Disease.
  • the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
  • the present invention includes a method of determining risk of an individual developing Alzheimer's disease, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive and optionally to negative control values, wherein levels of the biomarkers in the plasma sample of the individual that resemble the positive control and do not resemble the negative control are an indication that the individual is at risk of developing Alzheimer's disease.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine risk of an individual of developing Alzheimer's disease.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • the level of biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine risk of an individual of developing Alzheimer's disease.
  • the invention includes a method for monitoring Alzheimer's disease in an individual, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual at a first time, and comparing the level of the biomarkers in the plasma sample of the individual at the first time to a level of the biomarkers in a plasma sample of the individual at the second time.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to monitor Alzheimer's disease in the individual.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to monitor Alzheimer's disease in the individual.
  • the invention provides a method of assessing increased risk of developing Alzheimer's disease, the method comprising: obtaining a first sample of plasma from an individual blood at a first time, assessing the level of a biomarker for Alzheimer's disease in the first plasma sample to obtain a baseline level, obtaining a second sample of plasma from the individual at a second time to obtain a second level, assessing the level of the biomarker in the second plasma sample to obtain a second level, wherein if the second level is greater in the case of a biomarker that is over-expressed in AD or lower in the case of a biomarker that is under-expressed in AD, the individual is at an increased risk of developing Alzheimer's disease.
  • the biomarker is one or more biomarkers selected from Table 2.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
  • the invention includes a method of assessing the likelihood that a pharmaceutical agent is efficacious in treating Alzheimer's disease in an individual, the method comprising: obtaining a first sample of plasma from an individual in the absence of a pharmaceutical agent, assessing the level of a biomarker for Alzheimer's disease in the first plasma sample to obtain a baseline level, administering the pharmaceutical agent to the individual, obtaining a second sample of plasma from the individual after administration of the pharmaceutical agent, assessing the level of the biomarker for Alzheimer's disease in the second plasma sample to obtain a treated level, wherein the likelihood that the pharmaceutical agent treats Alzheimer's disease is increased if the treated level is lower than the baseline level in the case of a biomarker that is over- expressed in AD and higher than the baseline level in the case of a biomarker that is under-expressed in AD.
  • the biomarker is one or more biomarkers selected from Table 2.
  • the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to assess the likelihood that the pharmaceutical agent is efficacious in treating
  • the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess the likelihood that the pharmaceutical agent is efficacious in treating Alzheimer's disease in the individual.
  • the invention includes a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE provides a positive diagnosis of AD in the individual.
  • the invention includes a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides a positive diagnosis of AD in the individual.
  • the method further comprises conducting one or more cognitive tests on the individual to confirm the positive diagnosis of AD.
  • the method further comprises obtaining a CSF sample from the individual to confirm the positive diagnosis of AD.
  • the invention provides a method for screening to identify individuals at increased risk of developing AD, the method comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE provides an identification of an individual at increased risk of developing AD.
  • the invention provides a method for screening to identify individuals at increased risk of developing AD, the method comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and comparing the levels of each of the at least five biomarkers to non- AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides an identification of an individual at increased risk of developing AD.
  • the invention provides a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and determining whether, relative to non-AD controls, the levels of the at least five biomarkers provide a signature of a positive diagnosis of AD in the individual, wherein the signature comprises: a statistically significant increase in the levels of each of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE.
  • the invention provides a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and determining whether, relative to non-AD controls, the levels of the at least five biomarkers provide a signature of a positive diagnosis of AD in the individual, wherein the signature comprises: a statistically significant increase in the levels of each of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin.
  • the invention provides a kit for detecting Alzheimer's Disease in an individual with at least 80% sensitivity, comprising a reagent for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual.
  • the reagent is capable of detecting biomarkers of
  • Alzheimer's disease in a plasma sample Alzheimer's disease in a plasma sample.
  • the biomarkers of Alzheimer's disease comprise Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
  • the biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309.
  • the biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
  • Hepatocyte Growth Factor IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
  • Thrombomodulin tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
  • TRIP-1 tissue inhibitor of metalloproteinases-1
  • VCAM-1 vascular cell adhesion molecule- 1
  • VEGF Vascular endothelial growth factor
  • Von Willebrand Factor Von Willebrand Factor
  • the biomarkers include one or more biomarkers selected from Table 2.
  • the reagent is an antibody.
  • the reagent is on a solid support.
  • the kit further comprises an instruction manual.
  • Figure 1 is an image depicting Test set ROC curves for each model.
  • the solid black dot indicates the default cutoff of 0.50 and the square indicates the alternate cutoff to get sensitivity closest to 0.80.
  • PLS Partial Least Square Model
  • Figure 2 is a series of graphs depicting expression of analytes in CSF Autopsy Confirmed Samples. Graphs represent means and error bars represent SEM.
  • Figure 3 is a graph depicting ROC curves using logistic regression of baseline (age, gender and ApoE genotype) vs Rules-Based Medicine (RBM) analytes alone versus baseline plus 24 RBM analytes.
  • Age, gender, ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • alphal microglobulin e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • Apolipoprotein E e2/e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • alphal microglobulin e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • alphal microglobulin e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/
  • Osteopontin Pancreatic polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM, VEGF and von Willebrand Factor.
  • Figure 4A is an image demonstrating a correlation matrix analysis in the Alzheimer's disease samples exhibiting a high degree of correlation amongst the tested analytes.
  • Figure 4B is a chart demonstrating high correlation of the analytes with each other and that there is high correlation with other analytes on the panel.
  • NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway.
  • analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance.
  • Figure 5 is an image demonstrating that plasma levels of ApoE, CRP and IL-15 differ dependent upon genetic ApoE allele status independent of diagnosis. Levels of ApoE4 are lowest in subjects with one or more E4 allele(s) and highest in subjects with one or more E2 allele(s).
  • Figure 6 is an image depicting graphical summary of the training and test set estimates for sensitivity and specificity
  • Figure 7 is an image depicting test set sensitivity and specificity profiles across various cutoffs for each model.
  • Figure 8 is an image depicting the overall distribution of PLS variables importance scores.
  • Figure 9 is an image depicting the correlation matrix for the top 15 predictors.
  • Figure 10 is an image depicting the training set data points for the top 15 predictors.
  • the present invention provides a method for diagnosing, monitoring and/or staging neurological disorders comprising the use of a multivariate and/or univariate approach to identify a neurological disorder in an individual.
  • the present invention relates generally to diagnostic methods and markers, prognostic methods and markers, and therapy evaluators for neurodegenerative disorders, such as Alzheimer's Disease.
  • the markers of the invention are useful for detecting early stage Alzheimer's disease.
  • the method comprises the step of obtaining a sample of plasma from the individual's blood, and assessing the level of markers of Alzheimer's Disease in the plasma sample.
  • the present invention relates to markers of Alzheimer's Disease, methods for diagnosis of Alzheimer's Disease, methods of determining predisposition to Alzheimer's Disease, methods of monitoring
  • the invention further provides methods for permitting refinement of disease diagnosis, disease risk prediction, and clinical management of individuals associated with a neurodegenerative disorder.
  • Alzheimer's disease markers of the invention represent a plasma based panel for assessing Alzheimer's disease that can be used for determining the disease state or disease risk.
  • the detection of the selective markers of the invention in individuals, or samples obtained therefrom permits refinement of disease diagnosis, disease risk prediction, and clinical management of individuals being treated with agents that are associated with Alzheimer's disease.
  • the Alzheimer's disease markers of the invention include one or more of the markers shown in Table 2.
  • age, gender, and ApoE genotype are additional factors that are considered in identifying an individual for Alzheimer's disease.
  • markers of AD includes at least
  • markers of AD includes at least resistin, e3/e3, pancreatic polypeptide, e3/e4, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, and 1-309.
  • markers of AD includes at least
  • CXCL13 Chemokine (C-X-C motif) ligand 13 also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
  • Hepatocyte Growth Factor IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
  • the plasma based panel for assessing Alzheimer's disease comprises one or more of the following markers: Alpha-1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E- Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2,
  • Interleukin 10 Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases- 1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
  • TRIP-1 tissue inhibitor of metalloproteinases- 1
  • VCAM-1 vascular cell adhesion molecule- 1
  • VEGF Vascular endothelial growth factor
  • Von Willebrand Factor Von Willebrand Factor
  • age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
  • the invention includes a method comprising obtaining a sample from an individual, and assessing the level of one or more of Alpha-1 Microglobulin,
  • Angiopoietin-2 Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, and Von Willebrand Factor in the sample.
  • the invention should not be limited to only these markers disclosed herein (e.g., Table 2) because a skilled artisan when armed with the present disclosure would be able identify additional markers that can be used as indicators for Alzheimer's disease.
  • a test sample and a control sample can be subjected to any commercially available panel comprising a plurality of markers and analyzed according to the statistic models disclosed herein to identify markers associated with AD.
  • the disclosure presented here demonstrates a high degree of correlation amongst certain markers for identifying AD in an individual.
  • a skilled artisan when armed with the present disclosure, particularly Figures 4A and 4B, would recognize that analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance.
  • NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway.
  • markers that are associated with an AD specific pathophysiological pathway can be interchangeable. Accordingly, correlation amongst the markers of the invention provides means to identify other related markers associated with the specific pathophysiological pathway as being indicators for Alzheimer's disease.
  • biomarkers may be used in the methods disclosed herein. That is, the disclosed methods may include the determination of the level(s) of one biomarker, two or more biomarkers, three or more biomarkers, four or more biomarkers, five or more biomarkers, six or more biomarkers, seven or more biomarkers, eight or more biomarkers, nine or more biomarkers, ten or more biomarkers, fifteen or more biomarkers, etc., including a combination of all of the biomarkers in each or all of Tables 2, 3, 7, 8, 9, 10, 1 1, 12 or any fraction thereof.
  • the biomarkers are used in combination with other factors that predict for AD. Such factors include but are not limited to age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4).
  • an immunoassay is used for the assessment of a marker level.
  • a luminex technology multiplex immunoassay is used to assess the marker level.
  • a method of diagnosing Alzheimer's disease in an individual comprises the steps of obtaining a first sample of plasma from the individual at a first time; assessing the level of Alzheimer's disease markers using plasma based panel of the invention in the first plasma sample to obtain a baseline level; obtaining a second sample of plasma from the individual at a second time and assessing the level of
  • Alzheimer's disease markers in the second plasma sample to obtain a second level. If the second level is significantly altered compared to the baseline level, the individual is at an increased risk of developing or having Alzheimer's disease. In one embodiment, the second level is also compared to a reference population of an individual's without Alzheimer's disease; if the second level is significantly altered compared to the level derived from a reference population, the individual is at an increased risk of developing or having Alzheimer's disease.
  • the invention provides methods of monitoring a plasma panel of particular markers of Alzheimer's disease to evaluate the progress of a therapeutic treatment of Alzheimer's disease.
  • the invention provides methods for selecting a patient that is most likely to respond to treatment.
  • the invention also provides methods for screening an individual to determine if the individual is at increased risk of having Alzheimer's disease. Individuals found to be at increased risk can be given appropriate therapy and monitored using the methods of the invention.
  • an element means one element or more than one element.
  • “About” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ⁇ 20% or ⁇ 10%, more preferably ⁇ 5%, even more preferably ⁇ 1%, and still more preferably ⁇ 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • abnormal when used in the context of organisms, tissues, cells or components thereof, refers to those organisms, tissues, cells or components thereof that differ in at least one observable or detectable characteristic (e.g., age, treatment, time of day, etc.) from those organisms, tissues, cells or components thereof that display the "normal” (expected) respective characteristic. Characteristics which are normal or expected for one cell or tissue type, might be abnormal for a different cell or tissue type.
  • antibody refers to an immunoglobulin molecule which is able to specifically bind to a specific epitope on an antigen.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins.
  • the antibodies in the present invention may exist in a variety of forms including, for example, polyclonal antibodies, monoclonal antibodies, intracellular antibodies
  • synthetic antibody an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage as described herein.
  • the term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
  • the term “heavy chain antibody” or “heavy chain antibodies” comprises immunoglobulin molecules derived from camelid species, either by immunization with a peptide and subsequent isolation of sera, or by the cloning and expression of nucleic acid sequences encoding such antibodies.
  • the term “heavy chain antibody” or “heavy chain antibodies” further encompasses immunoglobulin molecules isolated from an animal with heavy chain disease, or prepared by the cloning and expression of VH (variable heavy chain immunoglobulin) genes from an animal.
  • an “immunoassay” refers to any binding assay that uses an antibody capable of binding specifically to a target molecule to detect and quantify the target molecule.
  • an antibody which recognizes an specific antigen, but does not substantially recognize or bind other molecules in a sample.
  • an antibody that specifically binds to an antigen from one species may also bind to that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific.
  • an antibody that specifically binds to an antigen may also bind to different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific.
  • the terms “specific binding” or “specifically binding”, can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • biomarker in the context of the present invention encompasses, without limitation, proteins, nucleic acids, and metabolites, together with their polymorphisms, mutations, variants, modifications, subunits, fragments, protein- ligand complexes, and degradation products, protein-ligand complexes, elements, related metabolites, and other analytes or sample-derived measures. Biomarkers can also include mutated proteins or mutated nucleic acids. Biomarkers also encompass non-blood borne factors or non-analyte physiological markers of health status, such as clinical parameters, as well as traditional laboratory risk factors. Biomarkers also include any calculated indices created mathematically or combinations of any one or more of the foregoing measurements, including temporal trends and differences.
  • the term “data” in relation to one or more biomarkers, or the term “biomarker data” generally refers to data reflective of the absolute and/or relative abundance (level) of a product of a biomarker in a sample.
  • the term “dataset” in relation to one or more biomarkers refers to a set of data representing levels of each of one or more biomarker products of a panel of biomarkers in a reference population of subjects.
  • a dataset can be used to generate a formula/classifier of the invention. According to one embodiment the dataset need not comprise data for each biomarker product of the panel for each individual of the reference population.
  • the "dataset" when used in the context of a dataset to be applied to a formula can refer to data representing levels of products of each biomarker for each individual in one or more reference populations, but as would be understood can also refer to data representing levels of products of each biomarker for 99%, 95%, 90%, 85%, 80%, 75%, 70% or less of the individuals in each of said one or more reference populations and can still be useful for purposes of applying to a formula.
  • “Differentially increased expression” or “up regulation” refers to biomarker product levels which are at least 10% or more, for example, 20%, 30%, 40%, or 50%, 60%, 70%, 80%, 90% higher or more, and/or 1.1 fold, 1.2 fold, 1.4 fold, 1.6 fold, 1.8 fold higher or more, than a control.
  • “Differentially decreased expression” or “down regulation” refers to biomarker product levels which are at least 10% or more, for example, 20%, 30%, 40%, or 50%, 60%, 70%, 80%, 90% lower or less, and/or 0.9 fold, 0.8 fold, 0.6 fold, 0.4 fold, 0.2 fold, 0.1 fold or less lower than a control.
  • a “formula,” “algorithm,” or “model” is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous or categorical inputs (herein called “parameters”) and calculates an output value, sometimes referred to as an "index” or “index value.”
  • “formulas” include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations.
  • Alzheimer's disease markers and other biomarkers are linear and non-linear equations and statistical classification analyses to determine the relationship between levels of Alzheimer's disease markers detected in a subject sample.
  • structural and synactic statistical classification algorithms, and methods of risk index construction utilizing pattern recognition features, including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Linear Discriminant Analysis (LDA), Support Vector Machines (SVM), Random Forest (RF), Partial Least Squares, Sparse Partial Least Squares, Flexible Discriminant Analysis, Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Nearest Shrunken Centroids (SC)", stepwise model selection procedures, Kth-Nearest Neighbor, Boosting or Boosted Tree, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others.
  • PCA Principal Components Analysis
  • LogReg Line
  • an “analyte”, as used herein refers to any substance or chemical constituent that is undergoing analysis.
  • an "analyte” can refer to any atom and/or molecule; including their complexes and fragment ions. The term may refer to a single component or a set of components. In the case of biological
  • molecules/macromolecules such analytes include but are not limited to: polypeptides, polynucleotides, proteins, peptides, antibodies, DNA, RNA, carbohydrates, steroids, and lipids, and any detectable moiety thereof, e.g. immunologically detectable fragments.
  • a “disease” is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate.
  • a “disorder” in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
  • Alzheimer's disease and “Alzheimer's disease” refer to a neurodegenerative disorder and encompass familial Alzheimer's disease and sporadic Alzheimer's disease.
  • familial Alzheimer's disease refers to familial Alzheimer's disease and sporadic Alzheimer's disease.
  • familial Alzheimer's disease refers to familial Alzheimer's disease and sporadic Alzheimer's disease.
  • Alzheimer's disease associated with genetic factors i.e., inheritance is demonstrated
  • sporadic Alzheimer's disease refers to Alzheimer's disease that is not associated with prior family history of the disease.
  • Symptoms indicative of Alzheimer's disease in human subjects typically include, but are not limited to, mild to severe dementia, progressive impairment of memory (ranging from mild forgetfulness to disorientation and severe memory loss), poor visual spatial skills, personality, changes, poor impulse control, poor judgment, distrust of others, increased stubbornness, restlessness, poor planning ability, poor decision making, and social withdrawal.
  • patients lose the ability to use language and communicate, and require assistance in personal hygiene, eating and dressing, and are eventually bedridden.
  • Hallmark pathologies within brain tissue include extracellular neuritic amyloid plaques, neurofibrillary tangles, neurofibrillary degeneration, granulovascular neuronal degeneration, synaptic loss, and extensive neuronal cell death.
  • “Increased risk of developing Alzheimer's disease” is used herein to refer to an increase in the likelihood or possibility of developing Alzheimer's disease. This risk can be assessed relative to an individual's own risk, or with respect to a reference population that does not have clinical evidence of Alzheimer's disease. The reference population may be representative of the individual with regard to approximate age, age group and/or gender.
  • Delaying development of Alzheimer's disease refers to a prolonging of the time to the development of Alzheimer's disease and/or delay in the rate of increased extent of Alzheimer's disease.
  • Alzheimer's disease refers to a decrease in the severity of Alzheimer's disease.
  • Alzheimer's patient As used herein, the terms "Alzheimer's patient”, “Alzheimer's disease patient”, and “individual diagnosed with Alzheimer's disease” all refer to an individual who has been diagnosed with Alzheimer's disease or has been given a probable diagnosis of Alzheimer's Disease.
  • An "individual with mild Alzheimer's disease” is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable Alzheimer's disease. In some instances, an "individual with mild Alzheimer's disease” has either been assessed with the Mini-Mental State Examination (MMSE) (referenced in Folstein et al, J. Psychiatr. Res 1975; 12: 1289-198) and scored 22-27 or would achieve a score of 22-27 upon MMSE testing.
  • MMSE Mini-Mental State Examination
  • An "individual with moderate Alzheimer's disease” is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable Alzheimer's disease. In some instances, an "individual with moderate Alzheimer's disease” has either been assessed with the MMSE and scored 16-21 or would achieve a score of 16-21 upon MMSE testing.
  • An "individual with severe Alzheimer's disease” is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable
  • Alzheimer's disease In some instances, an "individual with severe Alzheimer's disease" has either been assessed with the MMSE and scored 12-15 or would achieve a score of 12-15 upon MMSE testing.
  • the "level" of one or more biomarkers means the absolute or relative amount or concentration of the biomarker in the sample.
  • detection means assessing the presence, absence, quantity or amount (which can be an effective amount) of either a given substance within a clinical or subject-derived sample, including the derivation of qualitative or quantitative concentration levels of such substances, or otherwise evaluating the values or categorization of a subject's clinical parameters.
  • a “reference level” of a biomarker means a level of the biomarker that is indicative of a particular disease state, phenotype, or lack thereof, as well as combinations of disease states, phenotypes, or lack thereof.
  • a “positive" reference level of a biomarker means a level that is indicative of a particular disease state or phenotype.
  • a “negative” reference level of a biomarker means a level that is indicative of a lack of a particular disease state or phenotype.
  • an "Alzheimer's Disease-positive reference level" of a biomarker means a level of a biomarker that is indicative of a positive diagnosis of Alzheimer's Disease in a subject
  • an "Alzheimer's Disease-negative reference level" of a biomarker means a level of a biomarker that is indicative of a negative diagnosis of Alzheimer's Disease in a subject.
  • an "Alzheimer's Disease-positive reference level" of a biomarker means a level of a biomarker that is indicative of a positive diagnosis of Alzheimer's Disease in a subject
  • an "Alzheimer's Disease-negative reference level" of a biomarker means a level of a biomarker that is indicative of a negative diagnosis of Alzheimer's Disease in a subject.
  • an "Alzheimer's Disease-positive reference level" of a biomarker means a level of a biomarker that is indicative of a positive diagnosis of Alzheimer's Disease in a subject
  • Alzheimer's-Disease-progression-positive reference level of a biomarker means a level of a biomarker that is indicative of progression of Alzheimer's Disease in a subject
  • an "Alzheimer's-Disease-regression-positive reference level" of a biomarker means a level of a biomarker that is indicative of regression of the Alzheimer's Disease.
  • a “reference level" of a biomarker may be an absolute or relative amount or concentration of the biomarker, a presence or absence of the biomarker, a range of amount or concentration of the biomarker, a minimum and/or maximum amount or concentration of the biomarker, a mean amount or concentration of the biomarker, and/or a median amount or concentration of the biomarker; and, in addition, “reference levels” of combinations of biomarkers may also be ratios of absolute or relative amounts or concentrations of two or more biomarkers with respect to each other.
  • Appropriate positive and negative reference levels of biomarkers for a particular disease state, phenotype, or lack thereof may be determined by measuring levels of desired biomarkers in one or more appropriate subjects, and such reference levels may be tailored to specific populations of subjects (e.g., a reference level may be age-matched so that comparisons may be made between biomarker levels in samples from subjects of a certain age and reference levels for a particular disease state, phenotype, or lack thereof in a certain age group). Such reference levels may also be tailored to specific techniques that are used to measure levels of biomarkers in biological samples, where the levels of biomarkers may differ based on the specific technique that is used. Such reference levels may also be tailored to specific statistical models used to analyze levels of biomarkers in biological samples, where the levels of biomarkers may differ based on the specific statistical model that is used.
  • a "detector molecule” is a molecule that may be used to detect a compound of interest.
  • Non-limiting examples of a detector molecule are molecules that bind specifically to a compound of interest, such as, but not limited to, an antibody, a cognate receptor, and a small molecule.
  • an “effective amount” or “therapeutically effective amount” of a compound is that amount of compound which is sufficient to provide a beneficial effect to the individual to which the compound is administered.
  • An “effective amount” of a delivery vehicle is that amount sufficient to effectively bind or deliver a compound.
  • an "instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of a compound, composition, vector, or delivery system of the invention in the kit for effecting alleviation of the various diseases or disorders recited herein.
  • the instructional material can describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a individual.
  • the instructional material of the kit of the invention can, for example, be affixed to a container which contains the identified compound, composition, vector, or delivery system of the invention or be shipped together with a container which contains the identified compound, composition, vector, or delivery system.
  • the instructional material can be shipped separately from the container with the intention that the instructional material and the compound be used cooperatively by the recipient.
  • microarray refers broadly to both “DNA microarrays” and “DNA chip(s),” and encompasses all art-recognized solid supports, and all art-recognized methods for affixing nucleic acid molecules thereto or for synthesis of nucleic acids thereon.
  • patient refers to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein.
  • the patient, subject or individual is a human.
  • protein typically refers to large polypeptides.
  • sample or “biological sample” as used herein means a biological material isolated from an individual.
  • the biological sample may contain any biological material suitable for detecting the desired biomarkers, and may comprise cellular and/or non-cellular material obtained from the individual.
  • a “therapeutic” treatment is a treatment administered to an individual who exhibits signs of pathology, for the purpose of diminishing or eliminating those signs.
  • treat means reducing the frequency with which symptoms are experienced by an individual or subject or administering an agent or compound to reduce the frequency with which symptoms are experienced.
  • “treat” and “treating” are not limited to the case where the subject (e.g. patient) is cured and the disease is eradicated. Rather, the present invention also contemplates treatment that merely reduces symptoms, improves (to some degree) and/or delays disease progression.
  • treatment also refers to the alleviation, amelioration, and/or stabilization of symptoms, as well as delay in progression of symptoms of a particular disorder.
  • treatment of Alzheimer's disease includes any one or more of: elimination of one or more symptoms of Alzheimer's disease, reduction of one or more symptoms of Alzheimer's disease, stabilization of the symptoms of Alzheimer's disease (e.g., failure to progress to more advanced stages of Alzheimer's disease), and delay in progression (i.e., worsening) of one or more symptoms of Alzheimer's disease.
  • treating a disease or disorder means reducing the frequency with which a symptom of the disease or disorder is experienced by an individual.
  • Disease and disorder are used interchangeably herein.
  • ranges throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range and, when appropriate, partial integers of the numerical values within ranges. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range. Description
  • Alzheimer's disease is a progressive neurodegenerative disorder characterized by memory and cognitive impairments and other non-cognitive behavioral symptoms. Age is the strongest risk factor, wherein almost 50% of people over the age of 85 are affected. Early-onset Alzheimer's disease (EOAD) is associated with genetic mutations in amyloid precursor protein (APP), presenilin 1 (PSE 1) and presenilin 2
  • APP amyloid precursor protein
  • PSE 1 presenilin 1
  • presenilin 2 presenilin 2
  • LOAD sporadic or late-onset Alzheimer's disease
  • LOAD sporadic or late-onset Alzheimer's disease
  • genetic factors may account for as much as 80% of the disease risk associated with LOAD (Gatz et al. (2006) Arch. Gen. Psychiatry 63(2): 168-174).
  • monogenic mutations cause EOAD
  • the only extensively validated susceptibility gene for LOAD is the apolipoprotein E ( ⁇ - ⁇ 4) allele
  • the present invention provides novel Alzheimer's disease biomarkers present in a biological sample of an individual.
  • the biological sample is plasma.
  • the level of these biomarkers allow a more accurate diagnosis or prognosis of Alzheimer's disease in individuals that are at risk for Alzheimer's disease, that exhibit no clinical signs of Alzheimer's disease, or that exhibit minor clinical signs of Alzheimer's disease.
  • the measurement of the biomarkers of the invention allows the monitoring of Alzheimer's disease, such that a comparison of biomarker levels facilitates an evaluation of disease progression in individuals that have been diagnosed with Alzheimer's disease, or that do not yet exhibit any clinical signs of Alzheimer's disease.
  • the Alzheimer's disease biomarkers of the invention may be used in concert with known Alzheimer's disease biomarkers such that a more accurate diagnosis or prognosis of Alzheimer's disease may be made.
  • the invention provides a screening tool to detect Alzheimer's disease in an individual.
  • the screening tool includes a examining a multiplexed plasma panel to differentiate Alzheimer's disease from age-matched controls.
  • the screening tool encompasses a multivariate based approach combined a statistical model to predict Alzheimer's disease in an individual.
  • the multivariate approach useful in the invention encompasses the use of a multiplexed plasma panel in combination with age, gender and ApoE genetic status (6 co- variates e2/e2; e2/e3; e2/e4; e3/e4; e3/e3; e4/e4) for determining Alzheimer's disease in an individual.
  • the screening tool of the invention can differentiate Alzheimer's disease from age-matched controls with high sensitivity suitable for preliminary screening of individuals who might be eligible for clinical trial enrollment in early Alzheimer's disease studies.
  • the disclosure presented herein also includes additional markers associated with increased or decreased risk of Alzheimer's disease as shown in Table 2.
  • the present invention relates partly on the discovery of biomarkers that can differentiate Alzheimer's disease from controls and identify individuals at risk of progressing to dementia.
  • a two tiered strategy was employed to find the optimal discriminatory panel of biomarkers.
  • multivariate approaches were utilized to identify top plasma analytes that contributed to maximal differentiation using a training/test set approach.
  • plasma analytes were compared to expression in CSF from individuals with diagnosis confirmed by autopsy analysis.
  • a top list of analytes was identified based upon 1) performance in multivariate models, 2) expression in CSF from autopsy confirmed individuals and 3) upon linkage to disease by pathway analysis.
  • An objective was to determine whether a plasma based signature with a sensitivity of at least 80% could be achieved. In some instances, the objective was to determine whether a plasma based signature with a sensitivity of at least 80% and a specificity of at least 80% could be achieved.
  • a test with reasonable (at least 80%) sensitivity and specificity (Consensus report of the Working Group on:
  • a biomarker is an organic biomolecule which is differentially present in a sample taken from an individual of one phenotypic status (e.g., having a disease) as compared with an individual of another phenotypic status (e.g., not having the disease).
  • a biomarker is differentially present between the two individuals if the mean or median expression level of the biomarker in the different individuals is calculated to be statistically significant.
  • Biomarkers alone or in combination, provide measures of relative risk that an individual belongs to one phenotypic status or another. Therefore, they are useful as markers for diagnosis of disease, therapeutic effectiveness of a drug, and drug toxicity.
  • the invention provides methods for identifying one or more biomarkers which can be used to aid in the diagnosis, to diagnose, detect, predict neurological diseases such as neurodegenerative disorders.
  • the methods of the invention are carried out by obtaining a set of measured values for a plurality of biomarkers from a biological sample derived from a test individual, obtaining a set of measured values for a plurality of biomarkers from a biological sample derived from a control individual, comparing the measured values for each biomarker between the test and control sample, and identifying biomarkers which are significantly different between the test value and the control value also referred to as a reference value.
  • the process of comparing a measured value and a reference value can be carried out in any convenient manner appropriate to the type of measured value and reference value for the AD biomarker at issue.
  • “measuring” can be performed using quantitative or qualitative measurement techniques, and the mode of comparing a measured value and a reference value can vary depending on the
  • the levels may be compared by visually comparing the intensity of the colored reaction product, or by comparing data from densitometric or spectrometric measurements of the colored reaction product (e.g., comparing numerical data or graphical data, such as bar charts, derived from the measuring device).
  • data from densitometric or spectrometric measurements of the colored reaction product e.g., comparing numerical data or graphical data, such as bar charts, derived from the measuring device.
  • the measured values used in the methods of the invention will most commonly be quantitative values (e.g., quantitative
  • measured values are qualitative.
  • the comparison can be made by inspecting the numerical data, by inspecting representations of the data (e.g., inspecting graphical representations such as bar or line graphs).
  • a measured value is generally considered to be substantially equal to or greater than a reference value if it is at least about 95% of the value of the reference.
  • a measured value is considered less than a reference value if the measured value is less than about 95% of the reference value.
  • a measured value is considered more than a reference value if the measured value is at least more than about 5% greater than the reference value.
  • the process of comparing may be manual (such as visual inspection by the practitioner of the method) or it may be automated.
  • an assay device such as a luminometer for measuring chemiluminescent signals
  • a separate device e.g., a digital computer
  • Automated devices for comparison may include stored reference values for the AD biomarker(s) being measured, or they may compare the measured value(s) with reference values that are derived from contemporaneously measured reference samples.
  • the methods of the invention utilize "simple" or “binary" comparison between the measured level(s) and the reference level(s) (e.g., the comparison between a measured level and a reference level determines whether the measured level is higher or lower than the reference level).
  • a comparison showing that the measured value for the biomarker is altered from the reference value indicates or suggests a diagnosis of AD.
  • the process of comparing the measured values to identify indicators for disease state can be carried out using multiple marker analysis approach including but not limited to Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes,
  • FDA Flexible Discriminant Analysis
  • KNN K-Nearest Neighbors
  • Naive Bayes Naive Bayes
  • PLS Partial Least Squares
  • Random Forests Random Forests
  • Shrunken Centroids Sparse Partial Least Squares
  • Support Vector Machines approaches.
  • a biomarker is typically a protein, found in a bodily fluid, whose level varies with disease state and may be readily quantified. The quantified level may then be compared to a known value. The comparison may be used for several different purposes, including but not limited to, diagnosis of Alzheimer's disease, prognosis of Alzheimer's disease, and monitoring treatment of Alzheimer's disease.
  • biomarkers have been identified as associated with Alzheimer's disease.
  • biomarkers identified as being indicators of Alzheimer's disease include, but is not limited, to Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
  • Thrombomodulin TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, and any combination thereof.
  • age, gender, and ApoE genotype are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
  • the invention provides compositions for use in the methods of the invention.
  • the composition comprises a reagent that detects and/or quantitates an Alzheimer Disease biomarker.
  • the composition comprises a panel of reagents, each of which detects and/or quantitates a different Alzheimer Disease biomarker.
  • the Alzheimer Disease biomarker is an Alzheimer Disease biomarker shown in Table 2.
  • the composition comprises two or more reagents, each of which detects and/or quantitates a different one of two or more, three or more, four or more or five or more Alzheimer Disease biomarkers.
  • the compositions comprises a panel of reagents each of which detects and/or quantitates a different one of 24 Alzheimer Disease biomarkers shown in Table 2.
  • the composition comprises a panel of reagents for the detection and/or quantification of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
  • the composition comprises a panel of reagents for the detection and/or quantification of resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB,
  • the composition comprises a panel of reagents for the detection and/or quantification of Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vas
  • the combination of biomarkers associated with assessing Alzheimer's disease is collectively presented on a detectable medium referred to as a panel or plasma based panel.
  • a detectable medium referred to as a panel or plasma based panel.
  • Each of the biomarkers identified above may be used in concert with other biomarkers. For instance, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 10 or more, 15 or more, 20 or more, 30 or more, 40 or more, (and any integer value in between) Alzheimer's disease biomarkers may be used in concert with other, and with other known or to be known biomarkers for
  • the composition is an array of two or more detection reagents, e.g., antibodies and/or oligonucleotides each of which binds to a different one of two Alzheimer Disease biomarker proteins or nucleic acids, respectively.
  • the Alzheimer Disease biomarkers are proteins and the array contains antibodies sufficient to measure a statistically significant alteration in Alzheimer Disease biomarker expression compared to a reference value.
  • the invention provides a kit with at least two detection reagents, each of which binds to a different one of two Alzheimer Disease biomarker proteins, metabolites, or other analytes.
  • a. detection reagent is immobilized on a solid matrix such as a porous strip or bead to form at least one Alzheimer Disease biomarker detection site,
  • assessing cognitive function assessing cognitive impairment, diagnosing or aiding diagnosis of cognitive impairment by obtaining measured levels of one or more AD diagnosis biomarkers in a biological sample from an individual, such as for example, a plasma sample from an individual, and comparing those measured levels to reference levels.
  • the levels of Alzheimer's disease biomarkers of the invention may be assessed in several different biological samples, for example bodily fluids.
  • bodily fluids include whole blood, plasma, serum, bile, lymph, pleural fluid, semen, saliva, sweat, urine, and CSF.
  • the bodily fluid is selected from the group of whole blood, plasma, and serum.
  • the bodily fluid is whole blood.
  • the bodily fluid is plasma.
  • the bodily fluid is serum.
  • the bodily fluid is obtained from the individual using conventional methods in the art. For instance, one skilled in the art knows how to draw blood and how to process it in order to obtain serum and/or plasma for use in the method. Generally speaking, the method preferably maintains the integrity of the biomarkers of the invention such that it can be accurately quantified in the bodily fluid. Methods for collecting blood or fractions thereof are well known in the art. For example, see US Patent No. 5,286,262, which is hereby incorporated by reference in its entirety.
  • a bodily fluid may be obtained from any mammal known to suffer from Alzheimer's disease or that can be used as a disease model for Alzheimer's disease.
  • the mammal is a rodent. Examples of rodents include mice, rats, and guinea pigs.
  • the mammal is a primate. Examples of primates include monkeys, apes, and humans.
  • the mammal is a human.
  • the individual has no clinical signs of Alzheimer's disease.
  • the individual has mild clinical signs of Alzheimer's disease.
  • the individual may be at risk for Alzheimer's disease.
  • the individual has been diagnosed with Alzheimer's disease.
  • Assessment of biomarker levels may encompass assessment of the level of protein concentration or the level of enzymatic activity of the biomarker, wherever applies. In either case, the level is quantified such that a value, an average value, or a range of values is determined. In one embodiment, the level of protein concentration of the Alzheimer's disease biomarker is quantified.
  • kits include ProteoQwestTM Colohmetric Western Blotting Kits (Sigma-Aldrich, Co.), QuantiProTM bicinchoninic acid (BCA) Protein Assay Kit (Sigma-Aldrich, Co.), FluoroProfileTM Protein Quantification Kit (Sigma-Aldrich, Co.), the Coomassie Plus - The Better Bradford Assay (Pierce Biotechnology, Inc.), and the Modified Lowry Protein Assay Kit (Pierce Biotechnology, Inc.).
  • the protein concentration is measured using a luminex based multiplex immunoassay panel.
  • the invention should not be limited to any particular assay for assessing the level of a biomarker of the invention. That is, any currently known assay used to detect protein levels and assays to be discovered in the future can be used to detect the biomarkers of the invention.
  • assessing the level of a protein involves the use of a detector molecule for the biomarker.
  • Detector molecules can be obtained from commercial vendors or can be prepared using conventional methods available in the art.
  • Exemplary detector molecules include, but are not limited to, an antibody that binds specifically to the biomarker, a naturally-occurring cognate receptor, or functional domain thereof, for the biomarker, and a small molecule that binds specifically to the biomarker.
  • the level of a biomarker is assessed using an antibody.
  • non-limiting exemplary methods for assessing the level of a biomarker in a biological sample include various immunoassays, for example, immunohistochemistry assays, immunocytochemistry assays, ELISA, capture ELISA, sandwich assays, enzyme immunoassay, radioimmunoassay, fluorescent immunoassay, and the like, all of which are known to those of skill in the art. See e.g. Harlow et ah, 1988, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York; Harlow et ah, 1999, Using
  • Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY. The generation of polyclonal antibodies is accomplished by inoculating the desired animal with an antigen and isolating antibodies which specifically bind the antigen therefrom.
  • Monoclonal antibodies directed against one of the biomarkers identified herein may be prepared using any well known monoclonal antibody preparation procedures, such as those described, for example, in Harlow et al. (1988, In: Antibodies, A Laboratory Manual, Cold Spring Harbor, NY) and in Tuszynski et al. (1988, Blood, 72: 109-1 15). Human monoclonal antibodies may be prepared by the method described in U.S. patent publication 2003/0224490. Monoclonal antibodies directed against a biomarker such as EGF are generated from mice immunized with the biomarker using standard procedures as referenced herein.
  • a biomarker may be purified from a biological source that endogenously comprises the biomarker, or from a biological source recombinantly-engineered to produce or over-produce the biomarker, using conventional methods known in the art.
  • Exemplary nucleic acid and protein sequences for the eleven biomarkers described herein are readily available in public sequence databases, such as National Library of Medicine's genetic sequence database GenBank® (Benson et al, 2008, Nucleic Acids Research, 36(Database issue):D25-30).
  • GenBank® National Library of Medicine's genetic sequence database GenBank® (Benson et al, 2008, Nucleic Acids Research, 36(Database issue):D25-30).
  • antibodies are generated against the human homologs of each of the eleven biomarkers for practicing the methods using a biological sample from a human individual diagnosed with Parkinson's Disease.
  • Nucleic acid encoding the monoclonal antibody obtained using the procedures described herein may be cloned and sequenced using technology which is available in the art, and is described, for example, in Wright et al. (1992, Critical Rev. Immunol. 12(3,4): 125-168) and the references cited therein. Further, the antibody useful in the practice of the invention may be "humanized” using the technology described in Wright et al, (supra) and in the references cited therein, and in Gu et al. (1997,
  • a cDNA library is first obtained from mRNA which is isolated from cells, e.g., the hybridoma, which express the desired protein to be expressed on the phage surface, e.g., the desired antibody.
  • cDNA copies of the mRNA are produced using reverse transcriptase.
  • immunoglobulin fragments are obtained by PCR and the resulting DNA is cloned into a suitable bacteriophage vector to generate a bacteriophage DNA library comprising DNA specifying immunoglobulin genes.
  • a suitable bacteriophage vector to generate a bacteriophage DNA library comprising DNA specifying immunoglobulin genes.
  • the procedures for making a bacteriophage library comprising heterologous DNA are well known in the art and are described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY).
  • Bacteriophage which encode the desired antibody may be engineered such that the protein is displayed on the surface thereof in such a manner that it is available for binding to the antigen against which the antibody is directed.
  • Bacteriophage which express a specific antibody are incubated in the presence of the antigen, for instance, antigen immobilized on a resin or surface, the bacteriophage will bind to the antigen.
  • Bacteriophage which do not express the antibody will not bind to the antigen.
  • panning techniques are well known in the art and are described for example, in Wright et al, (supra).
  • a cDNA library is generated from mRNA obtained from a population of antibody -producing cells.
  • the mRNA encodes rearranged immunoglobulin genes and thus, the cDNA encodes the same.
  • Amplified cDNA is cloned into M13 expression vectors creating a library of phage which express human Fab fragments on their surface. Phage which display the antibody of interest are selected by antigen binding and are propagated in bacteria to produce soluble human Fab
  • Fab molecules comprise the entire Ig light chain, that is, they comprise both the variable and constant region of the light chain, but include only the variable region and first constant region domain (CHI) of the heavy chain.
  • Single chain antibody molecules comprise a single chain of protein comprising the Ig Fv fragment.
  • An Ig Fv fragment includes only the variable regions of the heavy and light chains of the antibody, having no constant region contained therein.
  • Phage libraries comprising scFv DNA may be generated following the procedures described in Marks et al, 1991, J. Mol. Biol.
  • phage display libraries in which the heavy and light chain variable regions may be synthesized such that they include nearly all possible specificities (Barbas, 1995, Nature Medicine 1 :837-839; de Kruif et al, 1995, J. Mol. Biol. 248:97-105) may also be used to prepare an antibody useful in the practice of the invention.
  • chromatography e.g., HPLC, gas chromatography, liquid chromatography
  • mass spectrometry e.g., MS, MS-MS
  • a chromatography medium comprising a cognate receptor for the biomarker or a small molecule that binds to the biomarker can be used to substantially isolate the biomarker from the biological sample.
  • Small molecules that bind specifically to a biomarker can be identified using conventional methods in the art, for instance, screening of compounds using combinatorial library methods known in the art, including biological libraries, spatially-addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the "one-bead one- compound” library method, and synthetic library methods using affinity chromatography selection.
  • the level of substantially isolated protein can be quantitated directly or indirectly using a conventional technique in the art such as spectrometry, Bradford protein assay, Lowry protein assay, biuret protein assay, or bicinchoninic acid protein assay, as well as immunodetection methods.
  • the level of enzymatic activity of the biomarker if such biomarker has an enzymatic activity maybe quantified.
  • enzyme activity may be measured by means known in the art, such as measurement of product formation, substrate degradation, or substrate concentration, at a selected point(s) or time(s) in the enzymatic reaction.
  • There are numerous known methods and kits for measuring enzyme activity For example, see US Patent No. 5,654, 152. Some methods may require purification of the Alzheimer's disease biomarker prior to measuring the enzymatic activity of the biomarker.
  • a pure biomarker constitutes at least about 90%, preferably, 95% and even more preferably, at least about 99% by weight of the total protein in a given sample.
  • Alzheimer's disease biomarkers of the invention may be purified according to methods known in the art, including, but not limited to, ion-exchange chromatography, size-exclusion chromatography, affinity chromatography, differential solubility, differential centrifugation, and HPLC. Determination of the Status of Alzheimer's Disease
  • the present invention is based on biomarker profiles or signatures determined for biological samples from individuals diagnosed with Alzheimer's Disease as well as from one or more other groups of control individuals (e.g., healthy control subjects not diagnosed with Alzheimer's Disease).
  • the profile for Alzheimer's Disease was compared to the profile for biological samples from the one or more other groups of control individual.
  • Those molecules differentially present, including those molecules differentially present at a level that is statistically significant, in the profile of Alzheimer's Disease samples as compared to another group (e.g., healthy control subjects not diagnosed with Alzheimer's Disease) were identified as biomarkers to distinguish those groups.
  • the biomarkers are discussed in more detail elsewhere herein.
  • AD biomarker levels relative to another reference level which may be relative to the level of another AD biomarker, may be obtained according to an appropriate statistical model disclosed herein.
  • high correlation amongst many biomarkers was observed to provide an indicator of disease state when analyzed using an appropriate statistical model.
  • the reference level used for comparison with the measured level for a AD biomarker may vary, depending on aspect of the invention being practiced, as will be understood from the foregoing discussion.
  • the "reference level” is typically a predetermined reference level, such as an average of levels obtained from a population that is not afflicted with AD, but in some instances, the reference level can be a mean or median level from a group of individuals including AD patients.
  • the predetermined reference level is derived from (e.g., is the mean or median of) levels obtained from an age-matched population.
  • the age-matched population comprises individuals with non-AD neurodegenerative disorders.
  • the reference level may be a historical reference level for the particular patient (e.g., a biomarker level that was obtained from a sample derived from the same individual, but at an earlier point in time).
  • the predetermined reference level is derived from (e.g., is the mean or median of) levels obtained from an age-matched population.
  • Age-matched populations are ideally the same age as the individual being tested, but approximately age-matched populations are also acceptable. Approximately age-matched populations may be within 1, 2, 3, 4, or 5 years of the age of the individual tested, or may be groups of different ages which encompass the age of the individual being tested. Approximately age-matched populations may be in 2, 3, 4, 5, 6, 7, 8, 9, or year increments (e.g. a "5 year increment" group which serves as the source for reference values for a 62 year old individual might include 58-62 year old individuals, 59-63 year old individuals, 60-64 year old individuals, 61-65 year old individuals, or 62-66 year old individuals.
  • the level(s) of the one or more biomarkers may be compared to
  • Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels using various techniques, including a simple comparison (e.g., a manual comparison) of the level(s) of the one or more biomarkers in the biological sample to Alzheimer's Disease- positive and/or Alzheimer's Disease-negative reference levels.
  • the level(s) of the one or more biomarkers in the biological sample may also be compared to Alzheimer's Disease- positive and/or Alzheimer's Disease-negative reference levels using one or more statistical analyses.
  • Statistical models useful in the present invention includes but are not limited to Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Na ' ive Bayes, Partial Least Squares (PLS), Random Forests, Shrunken Centroids, Sparse Partial Least Squares and Support Vector Machines approaches.
  • the statistical model is Partial Least Squares.
  • the AD biomarkers of the invention include one or more of the biomarkers shown in Table 2.
  • the combination of biomarkers associated with assessing Alzheimer's disease is collectively presented on a detectable medium referred to as a panel or plasma based panel.
  • Each of the biomarkers identified above may be used in concert with other biomarkers. For instance, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 10 or more, 15 or more, 20 or more, 30 or more, 40 or more, (and any integer value in between) Alzheimer's disease biomarkers may be used in concert with other, and with other known or to be known biomarkers for Alzheimer's disease.
  • age, gender, and ApoE genotype are additional factors that are considered in identifying an individual for Alzheimer's disease. Therefore, in some instances, predictors for AD in an individual are selected from the group consisting of Alpha- 1 Microglobulin,
  • Angiopoietin-2 Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), and any combination thereof.
  • ApoE genotype e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4
  • predictors of AD in an individual are selected from the group consisting of Resistin, e3/e3, Pancreatic Polypeptide, e3/e4, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, 1-309, and any combination thereof.
  • predictors of AD in an individual are selected from the group consisting of Cortisol, Pancreatic Polypeptide, osteopontin, IGF BP2, Resistin, and any combination thereof.
  • a plasma based signature for Alzheimer's disease can be detected in plasma and that signature can differentiate AD from healthy controls and other forms of dementia.
  • the signature for Alzheimer's disease includes the combination of biomarkers disclosed herein.
  • the signature for Alzheimer's disease is a combination of biomarkers and predictors of Alzheimer's disease disclosed herein.
  • the biomarkers of the invention in combination with other factors such as age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), can improve diagnostic and screening accuracy.
  • the biomarkers can also be combined with cognitive tests such as a simple memory test to improve diagnostic and screening accuracy.
  • the biomarkers of the invention can be combined with additional confirmatory CSF and imaging testing.
  • Tables 2 provide a listing of biomarkers that are useful for identifying an individual with AD.
  • certain biomarkers are increases and other biomarkers are decreased in AD compared to age-matched normal controls (Tables 10, 11, and 12.
  • a significant increase in a biomarker as compared to an appropriate control is indicative of AD
  • a significant decrease in a biomarker as compared to an appropriate control is indicative of AD.
  • correlation between biomarkers provide an indication of AD in an individual.
  • any one or more of the biomarkers listed in Table 2 can be used to identify AD in an individual as distinguished from other non-AD individuals.
  • the biomarkers of the invention can be used in diagnostic tests to assess the status of Alzheimer's disease in an individual, e.g., to diagnose Alzheimer's disease or to assess the degree of Alzheimer's disease in the individual.
  • the phrase "Alzheimer's disease status" includes any distinguishable manifestation of the disease, including non- Alzheimer's disease, e.g., normal or non-demented.
  • disease status includes, without limitation, the presence or absence of Alzheimer's disease (e.g., Alzheimer's disease v. non-Alzheimer's disease), the risk of developing disease, the stage of the disease, the progress of disease (e.g., progress of disease or remission of disease over time) and the effectiveness or response to treatment of disease. Based on this status, further procedures may be indicated, including additional diagnostic tests or therapeutic procedures or regimens.
  • ROC receiver operated characteristic
  • diagnostic tests that use the biomarkers of the invention exhibit a sensitivity and specificity of at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% and about 100%.
  • screening tools of the present invention exhibits a high sensitivity of at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% and about 100%.
  • screening tools should exhibit high sensitivity, but specificity can be low.
  • Diagnostics should have high sensitivity and specificity.
  • biomarkers While individual biomarkers are useful diagnostic biomarkers, it has been found that a combination of biomarkers can provide greater predictive value of a particular status than single biomarkers alone. Specifically, the detection of a plurality of biomarkers in a sample can increase the sensitivity and/or specificity of the test.
  • a combination of at least two biomarkers is sometimes referred to as a "biomarker profile” or “biomarker fingerprint.”
  • biomarker profile or “biomarker fingerprint.”
  • biomarker fingerprint A combination of at least two biomarkers.
  • Examples of Alzheimer's disease biomarkers of the invention include, but are not limited, to Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
  • Thrombomodulin TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, and any combination thereof.
  • age, gender, and ApoE genotype are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
  • the methods disclosed herein using the biomarkers listed in the tables presented herein may be used in combination with clinical diagnostic measures of Alzheimer's Disease and/or other neurodegenerative diseases. Combinations with clinical diagnostics may facilitate the disclosed methods, or confirm results of the disclosed methods (for example, facilitating or confirming diagnosis, monitoring progression or regression, and/or determining predisposition to Alzheimer's Disease).
  • Determining Alzheimer's disease status typically involves classifying an individual into one of two or more groups based on the results of the diagnostic test.
  • the diagnostic tests described herein can be used to classify an individual into a number of different states.
  • the invention provides methods for determining the presence or absence of Alzheimer's disease in an individual (status: Alzheimer's disease v. non-Alzheimer's disease).
  • the presence or absence of Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers in samples obtained from individuals and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular risk level.
  • the invention provides methods for determining the risk of developing disease in an individual.
  • Biomarker amounts or patterns are characteristic of various risk states, e.g., high, medium or low.
  • the risk of developing Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers in sample obtained from individuals and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular risk level.
  • the invention provides methods for determining the stage of Alzheimer's disease in an individual.
  • Each stage of the disease can be characterized by the amount of a biomarker or relative amounts of a set of biomarkers (i.e., a pattern) that are found in a sample obtained from the individual.
  • the stage of Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular stage.
  • the invention provides methods for determining the course of Alzheimer's disease in an individual.
  • Disease course refers to changes in disease status over time, including disease progression (worsening) and disease regression (improvement).
  • the amounts or relative amounts (e.g., the pattern) of the biomarkers changes.
  • levels of various biomarkers of the present invention increase with progression of disease.
  • this method involves measuring the level of one or more biomarkers in an individual at two or more different time points, e.g., a first time and a second time, and comparing the change in amounts. The course of disease is determined based on these comparisons.
  • the levels of various biomarkers of the invention decreases with disease progression.
  • the level of one or more biomarkers in a sample from an individual is measured at two or more different time points, e.g., a first time and a second time, and the change in levels, if any is assessed. The course of disease is determined based on these comparisons.
  • changes in the rate of disease progression may be monitored by measuring the level of one or more biomarkers at different times and calculating the rate of change in biomarker levels.
  • the ability to measure disease state or rate of disease progression is important for drug treatment studies where the goal is to slow down or arrest disease progression using therapy.
  • Additional embodiments of the invention relate to the communication of the results or diagnoses or both to technicians, physicians or patients, for example.
  • computers are used to communicate results or diagnoses or both to interested parties, e.g., physicians and their patients.
  • the methods of the invention further comprise managing individual treatment based on their disease status.
  • Such management includes the actions of the physician or clinician subsequent to determining Alzheimer's disease status. For example, if a physician makes a diagnosis of Alzheimer's disease, then a certain regime of treatment, such as prescription or administration of the therapeutic compound might follow. Alternatively, a diagnosis of non-Alzheimer's disease might be followed by further testing to determine any other diseases that might the patient might be suffering from. Also, if the test is inconclusive with respect to Alzheimer's disease status, further tests may be called for.
  • a diagnosis based on the presence or absence or relative levels in the biological sample of an individual of the relevant biomarkers disclosed herein is communicated to the individual as soon as possible after the diagnosis is obtained.
  • the present invention provides a method of assessing efficacy of a treatment of Alzheimer's disease in a patient comprising: a) determining a baseline level of biomarkers in a first sample obtained from the patient before receiving the treatment; b) determining the level of same biomarkers in a second sample obtained from the patient after receiving the treatment; wherein an alteration in the levels of the biomarkers in the post-treatment sample is correlated with a positive treatment outcome.
  • the experiments disclosed herein were designed to develop an assay to identify universal accepted biomarkers for diagnosing, screening, monitoring and staging neurodegenerative diseases such as Alzheimer's disease that are fast, more accurate, and less expensive.
  • the results presented herein demonstrate that a diagnostic assay has been developed that can detect among other things early onset of Alzheimer's disease.
  • Detection of early onset of Alzheimer's disease is believed to increase the success rate of the individual being successfully treated for Alzheimer's disease.
  • the diagnostic method of the present invention can be applied to subjects who have been previously diagnosed with Alzheimer's disease, those who are suspected of having Alzheimer's disease, and those at risk of developing Alzheimer's disease.
  • patients diagnosed with dementia in particular, those patients who were previously clinically normal, are suitable subjects.
  • the present invention be limited to use with any particular subject types.
  • the subject is a human subject.
  • the subject is selected from the group consisting of subjects displaying pathology resulting from Alzheimer's disease, subjects suspected of displaying pathology resulting from Alzheimer's disease, and subjects at risk of displaying pathology resulting from Alzheimer's disease.
  • the Alzheimer's disease diagnosed using the method of the present invention is selected from the group consisting of late onset Alzheimer's disease, early onset Alzheimer's disease, familial Alzheimer's disease and sporadic Alzheimer's disease.
  • EOAD Early-onset Alzheimer's disease
  • EOAD is a rare form of Alzheimer's disease in which individuals are diagnosed with the disease before age 65. Less than 10% of all Alzheimer's disease patients have EOAD. Younger individuals who develop Alzheimer's disease exhibit more of the brain abnormalities that are normally associated with Alzheimer's disease.
  • EOAD is usually familial and follows an autosomal dominant inheritance pattern.
  • mutations in three genes including amyloid precursor protein (APP) on chromosome 21, presenilin 1 (PSEN1) on chromosome 14 and presenilin 2 (PSEN2) on chromosome 1 have been identified in families with EOAD . Mutations in the APP, PSEN1 and PSEN2 genes account for about 50% of the disease .
  • APP amyloid precursor protein
  • PSEN1 presenilin 1
  • PSEN2 presenilin 2
  • Late-onset Alzheimer's disease is the most common form of Alzheimer's disease, accounting for about 90% of cases and usually occurring after age 65.
  • LOAD strikes almost half of all individuals over the age of 85 and may or may not be hereditary. It is a complex and multifactorial disease with the possible involvement of several genes. Genome-wide linkage or linkage disequilibrium studies on LOAD have provided informative data for the existence of multiple putative genes for Alzheimer's disease on several chromosomes, with the strongest evidence on chromosomes 12, 10, 9 and 6. LOAD cases tend to be sporadic, wherein there is no family history of the disease. Genetic susceptibility at multiple genes and interaction between these genes as well as environmental factors are most likely responsible for the etiology of LOAD.
  • Twin data on incident cases indicates that almost 80% of the LOAD risk is attributable to genetic factors.
  • the Apolipoprotein E (APOE) gene on chromosome 19ql3 has been identified as a strong risk factor for LOAD.
  • the ⁇ - ⁇ 4 allele has been established as a strong susceptibility marker that accounts for nearly 30% of the risk in late-onset Alzheimer's disease.
  • three variants of APOE, encoded by codons 1 12 and 158, have been found to modify the risk of LOAD.
  • the effect of the ⁇ - ⁇ 4 allele is dose related, wherein one or two copies of the ⁇ - ⁇ 4 allele are associated with 3-fold or 15-fold risk, respectively.
  • the effect of the ⁇ - ⁇ 4 allele on Alzheimer's disease risk appears to decline with increasing age (M. Ilyas Kamboh (2004), supra).
  • a profile of biomarkers for Alzheimer's disease can be detected in plasma and that profile can differentiate AD from healthy controls and other forms of dementia.
  • the profiles for Alzheimer's disease includes the biomarkers disclosed herein.
  • the profile for Alzheimer's disease is a combination of biomarkers and other factors of Alzheimer's disease disclosed herein.
  • the biomarkers of the invention in combination with other factors such as age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), can improve diagnostic and screening accuracy.
  • the biomarkers can also be combined with cognitive tests such as a simple memory test to improve diagnostic and screening accuracy.
  • the biomarkers of the invention can be combined with additional confirmatory CSF and imaging testing.
  • biomarkers of the invention can be combined with existing criteria for dementia to improve diagnostic and screening accuracy of
  • Dementia is the decline of memory and other cognitive functions in comparison with the patient's previous level of function as determined by a history of decline in performance and by abnormalities noted from clinical examination and neuropsychological tests.
  • a diagnosis of dementia cannot be made when consciousness is impaired by delirium, drowsiness, stupor, or coma or when other clinical abnormalities prevent adequate evaluation of mental status.
  • Dementia is a diagnosis based on behavior and cannot be determined by computerized tomography, electroencephalography, or other laboratory instructions, although specific causes of dementia may be identified by these means.
  • the biomarkers of the invention can be combined with existing criteria Alzheimer's disease.
  • a clinical diagnosis of probable Alzheimer's disease can be made with confidence if there is a typical insidious onset of dementia with progression and if there are no other systemic or brain diseases that could account for the progressive memory and other cognitive deficits.
  • disorders that must be excluded are manic depressive disorder, Parkinson's disease, multiinfarct dementia, and drug intoxication; less commonly encountered disorders that may cause dementia include thyroid disease, pernicious anemia, luetic brain disease and other chronic infections of the nervous system, subdural hematoma, occult hydrocephalus, Huntington's disease, Creutzfeldt- Jakob disease, and brain tumors.
  • a diagnosis of definite Alzheimer's disease requires histopathologic confirmation.
  • a clinical diagnosis of possible Alzheimer's disease may be made in the presence of other significant diseases, particularly if, on clinical judgment, Alzheimer's disease is considered the more likely cause of the progressive dementia.
  • the clinical diagnosis of possible rather than probable Alzheimer's disease may be used if the presentation or course is somewhat aberrant.
  • the information needed to apply these criteria is obtained by standard methods of examination: the medical history; neurologic; psychiatric, and clinical examinations; neuropsychological tests; and laboratory studies.
  • biomarkers for Alzheimer's Disease described herein may also be biomarkers for neurodegenerative diseases in general.
  • Alzheimer's Disease biomarkers may be used in the methods described herein for neurodegenerative diseases in general. That is, the methods described herein with respect to Alzheimer's Disease may also be used for diagnosing (or aiding in the diagnosis of) a neurodegenerative disease, methods of monitoring progression/regression of a neurodegenerative disease, methods of assessing efficacy of compositions for treating a neurodegenerative disease, methods of screening a composition for activity in modulating biomarkers associated with a neurodegenerative disease, methods of identifying potential drug targets for neurodegenerative diseases, and methods of treating a neurodegenerative disease. Such methods could be conducted as described herein with respect to Alzheimer's Disease.
  • kits are envisaged for every method disclosed.
  • the following description of a kit useful for diagnosing Alzheimer's disease in an individual by measuring the level of a biomarker in a biological sample therefore is not intended to be limiting and should not be construed that way.
  • the kit may comprise a negative control containing a biomarker at a concentration of about the concentration of the biomarker which is present in a biological sample of an individual who does not have Alzheimer's disease or does not have increased risk for Alzheimer's disease.
  • the kit may also include a positive control containing the biomarker at a concentration of about the concentration of the biomarker which is present in a biological sample of an individual who as Alzheimer's disease or has increased risk for Alzheimer's disease.
  • the kit includes a panel of biomarkers including one or more of the biomarkers shown in Table 2.
  • the invention should not be limited to only these markers disclosed herein (e.g., Table 2) because a skilled artisan when armed with the present disclosure would be able identify additional markers that can be used as indicators for Alzheimer's disease.
  • a test sample and a control sample can be subjected to any commercially available panel comprising a plurality of markers and analyzed according to the statistic models disclosed herein to identify markers associated with AD.
  • NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway.
  • analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance.
  • markers that are associated with an AD specific pathophysiological pathway can be interchangeable. Accordingly, correlation amongst the markers of the invention provides means to identify other related markers associated with the specific pathophysiological pathway as being indicators for
  • biomarkers in each or all of Tables 2, 3, 7, 8, 9, 10, 11, 12 or any fraction thereof can be included in the kit.
  • other factors that predict for of AD can be included in the kit. Such factors include but are not limited to ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4).
  • the kit comprises a panel comprising at least Cortisol, Pancreatic Polypeptide, osteopontin, IGF BP2, and Resistin.
  • the kit comprises a panel comprising at least Resistin, e3/e3, Pancreatic Polypeptide, e3/e4, ApoD, G-CSF, MlP lbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, 1-309.
  • the kit comprises a panel comprising Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, and Von Willebrand Factor.
  • the kit comprises reagents to assess age, gender,
  • ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual.
  • the kit of the invention can be used to assess the status of Alzheimer's disease in an individual, e.g., to diagnose Alzheimer's disease or to assess the degree of Alzheimer's disease in the individual.
  • the phrase "Alzheimer's disease status" includes any distinguishable manifestation of the disease, including non-Alzheimer's disease, e.g., normal or non-demented.
  • disease status includes, without limitation, the presence or absence of Alzheimer's disease (e.g., Alzheimer's disease v. non-Alzheimer's disease), the risk of developing disease, the stage of the disease, the progress of disease (e.g., progress of disease or remission of disease over time) and the effectiveness or response to treatment of disease. Based on this status, further procedures may be indicated, including additional diagnostic tests or therapeutic procedures or regimens.
  • the kit includes an instructional material for use in the diagnosis of Alzheimer's disease in an individual.
  • the instructional material can be a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the method of the invention in the kit for assessment of Alzheimer's disease risk in a individual.
  • the instructional material of the kit of the invention may, for example, be affixed to a container which contains other contents of the kit, or be shipped together with a container which contains the kit. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the contents of the kit be used cooperatively by the recipient.
  • the experiments disclosed herein were designed to develop an assay to identify universal accepted biomarkers for diagnosing, monitoring and staging neurodegenerative diseases such as Alzheimer's disease that are fast, more accurate, and less expensive.
  • the results presented herein demonstrate that a diagnostic assay has been developed that can detect among other things early onset of Alzheimer's disease.
  • Example 1 Assessing diagnostic accuracy of a plasma based multiplexed immunoassay panel to identify Alzheimer's disease
  • prodromal Alzheimer's can be identified through the use of cognitive tests combined with cerebrospinal fluid (CSF) and imaging biomarkers.
  • CSF cerebrospinal fluid
  • Alzheimer's disease The present study examines the suitability of a multiplexed plasma panel as a screening tool to differentiate Alzheimer's disease from age-matched controls.
  • a luminex based multiplex immunoassay panel consisting of 190 analytes was utilized in plasma samples derived from the University of Pennsylvania.
  • the strategy to identify a plasma based panel included multivariate based approaches combined with examining changes in both CSF in both autopsy confirmed and non- autopsy confirmed individuals from the UPenn biobank. Final analytes were chosen based upon high performance in multivariate models and upon expression in CSF from autopsy confirmed individuals.
  • Multivariate training/test set models using all 190 analytes suggested Alzheimer's disease individuals could be differentiated from controls using analytes on the panel. Partial least square and shrunked centroid algorithms exhibited the best performance. Many of the top analytes identified in the multivariate and ANOVA analysis were also expressed in CSF obtained from autopsy confirmed individuals.
  • Multivariate and CSF results were combined to generate a final panel consisting of 24 multiplex analytes plus age, gender and ApoE genetic status (6 co-variates e2/e2; e2/e3; e2/e4; e3/e4; e3/e3; e4/e4).
  • Performance of the panel in differentiating Alzheimer's disease from controls in the entire UPenn sample set using a logistic regression analysis exhibited 80% sensitivity 80% specificity.
  • Multivariate approaches combined with CSF-plasma expression and pathway analysis were utilized to identify a 32 variate algorithm that differentiates Alzheimer's disease from age-matched controls with high sensitivity suitable for preliminary screening of individuals who might be eligible for clinical trial enrollment in early Alzheimer's disease studies.
  • Luminex xMAP panel was used for genotyping APOE nucleotides 334 T/C and 472 C/T with an ABI 7900 real time thermocylcer using DNA freshly prepared from EDTA whole blood.
  • Luminex xMAP panel was used for genotyping APOE nucleotides 334 T/C and 472 C/T with an ABI 7900 real time thermocylcer using DNA freshly prepared from EDTA whole blood.
  • the luminex technology multiplexes immunoassays on the surface of polystyrene microsphere beads.
  • the microsphere beads are loaded with a ratio of two spectrally distinct flourochromes yielding up to 100 uniquely color-coded beads.
  • the beads are coated with capture antibodies specific for the assay and run in either standard sandwich or competitive immunoassay format. Capture-antibody microspheres are incubated with blocking solution and diluted plasma sample or calibration controls for one hour. Beads are rinsed and biotinylated detection reagent added.
  • Streptavidin-phycoerthyrin is then added to each well and incubated for 60 minutes. Following wash steps, beads are resuspended in reading solution and read on the luminex instrument. Details of each assay procedure are provided by RBM in validation reports that are compliant with CLIA standards. Some of the assays have defined a lower limit of quantitation. For the purposes of the present study, the lower limit of detection was utilized. Validation details of all the assays are available from Rules Based Medicine (http://www.rulesbasedmediciiie.com). Plasma Collection Methods
  • analytes that had more than 10% missing values were excluded from the analysis. Values that were reported as LOW (below the lowest assay limit which is defined by RBM as a value below the lowest calibrator for the individual assay) were imputed by taking the reported low detection limit for individual assays (LDD) and dividing by 2. Individual analyte distributions were tested for normalcy using Box-Cox analysis. If necessary, analytes were log transformed and checked again for normalcy. The majority of analytes exhibited non-normal distributions and were log transformed. For purposes of the univariate analysis, multidimensional scaling and Mahalanobis distances were used to detect outliers.
  • Outliers were defined as 5 standard deviations beyond the mean and were replaced with the 5 STD value. A major concern in the analysis was control of type I error rate due to relatively large number of plasma proteins in the multiplex panel. False discovery (FDR) corrections were applied to p-values to account for the multiple comparisons. ANOVA models including diagnosis, age, gender and ApoE4 allele status were utilized in the final model. ApoE allele status was classified into 6 subcategories; e2/e2, e2/e3, e2/e4, e3/e3, e3/e4, e4/e4.
  • Table 1 summarizes demographic characteristics of the UPenn sample set. Two hundred and sixty six plasma samples were shipped for multiplex analysis. Of the 266, 92 were from Alzheimer's disease, 126 were from healthy elderly controls and 48 were from individuals with other forms of dementia. Control and dementia individuals from the UPenn cohort were significantly younger than Alzheimer's disease. Thus age was included as a co-variate in all models. Amongst the groups, females were more highly represented, but there were no significant differences in gender ratios between the groups. Gender was also included as a co-variate in all the models. The prevalence of ApoE4 allele in the Alzheimer's disease group was 63%, very similar to what was observed in general Alzheimer's disease population.
  • Table 2 summarizes all the analytes included in the RBM panel. Light gray highlights those analytes that had more than 10% missing or had more than 10% LOW values. Multivariate approaches were combined with CSF expression patterns for feature selection. The top 24 performers are highlighted in dark gray (Table 2).
  • the multivariate analyses in this context are the predictive models that were performed (e.g. Random forest, PLC, etc.) for the goal of predicting the outcome.
  • Table 2 Analyte and final model summary. Analytes with more than 10% missing or had more than 10% LOW values (highlighted in light gray). Final panel (highlighted in dark grey) were selected based upon multivariate results and upon expression from auto sy confirmed individuals.
  • Multivariate strategies included a shrunken centroid algorithm developed for predictive analysis of microarrays (PAM) (Tusher et al, 2001 Proc Natl Acad Sci U S A. 98(9):5116-5121), FDA, KNN, logistic regression, Naive Bayes, partial least squares, random forest, sparse partial least square, boosted trees, linear discriminant analysis (data not shown), principal components (data not shown) and support vector machine (Vapnik et al, 1999 IEEE Trans Neural Netw. 10(5):988-999). ROC curves from the analysis are shown in Figure 1.
  • Table 3 Summary of analyte levels in UPenn Plasma and ANOVA Results: Age and gender included as co-variates and p values reported for Alzheimer's disease v control
  • Alzheimer' s disease 0.0054 Thrombospon Alzheimer's disease 0.0491 ⁇ ng ml) 0.53 + 0.25 din- 1 (ng.ml) 293.2 + 11228.5 ⁇ 0.0001*
  • a logistic regression model was utilized to compare performance of a baseline model (age, gender and ApoE allele status) to a model including only the 24 top plasma analytes and a model including all 24 plasma analytes plus the baseline model. All models were benchmarked at 80% sensitivity to allow comparisons across the models. In general, the baseline demographic model using only age, gender and ApoE4 allele showed positive predictive value of 52%. A model including all 24 analytes showed a positive predictive value of 64% and a model including all 24 analytes and baseline demographics showed a reasonable positive predictive value of 75% (see Table 4). ROC curves for each of the models are depicted in Figure 3.
  • the final logistic regression model including age, gender, ApoE allele and 24 plasma analytes yielded a 80% sensitivity and 80% specificity in differentiating Alzheimer's disease from controls. Model showed very good performance in differentiating Alzheimer's disease from other forms of dementia as well.
  • a model was attempted using 12 of the 18 analytes described by Ray et al. (2007 Nat Med. 13(1 1): 1359-1362) (Angiopoietin-2, EGF, G-CSF, ICAM- 1, Interleukin 3, Interleukin 8/CXCL8, M-CSF, PDGF-BB, PARC/CCL18,
  • FIG. 4A Correlation matrix analysis in the Alzheimer's disease samples showed a high degree of correlation amongst the 24 analytes (Figure 4A).
  • Figure 4B is a chart also demonstrating high correlation of the analytes with each other and that there is high correlation with other analytes on the panel.
  • any of the individual 24 analyte can be switched out with any analyte that showed significant correlation with another analyte on the panel (p ⁇ 0.05).
  • NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an Alzheimer's disease specific pathophysiological pathway.
  • Many of the analytes in the 190 panel showed high correlation and it was possible to interchange the top 24 analytes with other highly correlated analytes and obtain similar differentiation performance (data not shown).
  • Figure 5 compares apolipoprotein E levels in individuals as a function of their ApoE genotype.
  • individuals with either an e3/e4 or e4/e4 genotype showed lower apolipoprotein levels compared to individuals with e2/e2, e3/e2 or e3/e3 genotypes.
  • Individuals with an e2/e4 genotype exhibited levels more closely matched to e3 and e2 groups. Although the numbers are very small, individuals homozygous for E2 exhibited much higher protein levels.
  • Alzheimer's disease individuals were present in each of the ApoE genetic groups with the exception of e2 homozygotes.
  • Alzheimer's disease individuals prior to the onset of dementia will be critical for successful development of Alzheimer's disease disease modifying drug.
  • CSF and imaging research tools currently available to identify individuals at risk of progressing to dementia, these tools are difficult to implement as screening tools for individual enrollment due to the cost and invasive nature of CSF testing.
  • a simple blood test would aid in identifying those eligible for more confirmatory biomarker and cognitive testing and would help raise awareness around dementia prevention.
  • the top lists of obtained from multivariate approaches were compared to ANOVA lists and lists obtained from examining expression of analytes in CSF from autopsy confirmed individuals.
  • a final model containing 24 analytes was selected for further analysis. Inclusion of the 24 multiplex analytes improved performance over baseline demographic models in identifying individuals with Alzheimer's disease. Logistic regression estimates of model performance showed 80% sensitivity and 80% specificity in differentiating Alzheimer's disease from controls. However, caution should be exercised as these estimates are likely to degrade in a population setting and when tested in independent datasets.
  • Correlation analysis utilized in the current study to identify analyte redundancy in the panel was actually quite revealing as many of the analytes showed strong correlations. The finding may be indicative of a biological signaling cascade activated during pathological sequeale associated with dementia. Further pathway analysis may shed insight into the underlying pathophysiological and specific endophenotypes associated with Alzheimer's disease and other forms dementia.
  • Apolipoprotein E is a glycoprotein involved in lipid transport and a ligand for receptor mediated endocytosis. It is one of the major high density lipid (HDL) components in brain and believed to be critical for cholesterol transport involved in brain synaptic turnover.
  • ApoE is the only gene consistently shown to increase the risk of late onset Alzheimer's disease and E4 is believed to confer greater risk while E2 is believed to be protective for dementia.
  • a novel genetic risk factor has been identified in another apolipoprotein, ApoJ also known as clusterin. The current study did not identify any changes in ApoJ levels in either cohort and additional studies in CSF are needed to further understand relevance.
  • the current findings support that a plasma based signature for Alzheimer's disease can be detected in plasma and that signature can differentiate Alzheimer's disease from healthy controls and other forms of dementia.
  • the signature can improve diagnostic accuracy over age and ApoE4 allele status alone, but does not rise to the level of a standalone diagnostic tool.
  • the panel may, when combined with a simple memory test, have utility as a screening tool for individuals with a cognitive complaint who would then be eligible for more confirmatory CSF and imaging testing.
  • the data set consisted of 165 characteristics (i.e. variables) collected on
  • Random Forests A sequence of 3 values for the number of retained variables were used to tune the model (ranging from 2 to 2). Breiman. Random forests. Machine learning (2001) vol. 45 (l) pp. 5-32.
  • Boosted Tree candidate models had interaction depths raining from 2 to 10, while the number of boosting iterations ranged from 100 to 2000. Friedman. Greedy function approximation: a gradient boosting machine. Annals of Statistics (2001) pp. 1 189-1232.
  • ⁇ Naive Bayes models were computed using a Gaussian kernel or a non-parametric density estimate.
  • Nearest Shrunken Centroids candidate models used threshold parameters ranging from 0.092 to 2.583.
  • Nearest Shrunken Centroids Tibshirani et al. Class prediction by nearest shrunken centroids, with applications to DNA microarrays. Statistical Science (2003) pp. 104-117.
  • Support Vector Machines a radial basis function was used with cost parameter values ranging from 0.1 to 100.
  • the tuning parameter used 50 iterations of the bootstrap to compute resampled estimates of the sensitivity and specificity. Previous modeling for these data showed that high specificity can be easily attained, so the final model was chosen by maximizing the sensitivity value across the candidate models.
  • Sensitivity and specificity were used to evaluate the data on the training and test sets. On the training set, 50 iterations of the bootstrap were used to get reasonable estimates of performance and to differentiate models. Sensitivity, specificity and the area under the ROC curve were also calculated on the 43 samples in the test set. Table 5 shows the results for the training data across all the models. Table 6 also contains the test set results. Figure 6 shows these results in graphical terms. Table 5: Training set resampling results across various models
  • Naivsj B& is 0.611 I i t
  • Figure 1 shows the ROC curves for the test set with each model.
  • the solid black point shows the sensitivity/specificity combination that corresponds to using a cutoff of 0.50 on the probability estimate of being Alzheimer's disease.
  • the solid square indicates an alternate choice based on getting the sensitivity as close as possible.
  • the test in each panel shows the alternative cutoff and the corresponding sensitivity/specificity estimates.
  • Figure 7 also shows the sensitivity and specificity profiles across various probability cutoffs. This plot can demonstrate how adaptable each model is to alternative cutoffs. It was observed from the models that were run on these data, the partial least squares (PLS) model appeared to have the best combination of sensitivity and specificity, as well as a high test set ROC AUC value. The PLS model used all the predictors in the data set.
  • PLS partial least squares
  • Table 7 has a list of the top 15 predictors that contribute to the model. The overall distribution of the scores is shown in Figure 8. In Figure 8, there is not clear cluster of variables with "high” and “low” importance. The rankings of the genotype variables were not in the top 15.
  • Table 7 The top 15 most important predictors for the PLS model (from most important to least)
  • Figure 9 shows the correlation patterns between the variables and Figure 8 shows the distribution of the training set data for each predictor.
  • Figure 10 shows no single predictor that significantly differentiates Alzheimer's disease from control. This also supports the idea that the models require a large number of predictors to achieve adequate performance.
  • ANCOVA Alzheimer's disease vs Control only
  • DIAG 1 1 0.09757082 4 .9286 0.0275
  • DIAG 1 1 0.05191313 0.6177 0.4328
  • DIAG 1 1 0.00742880 0.2975 0.5860
  • DIAG 1 1 0.00001020 0.0008
  • Gender 1 1 0.63626015 20.1505 ⁇ .0001
  • DIAG 1 1 0.0021 4 321 0. 44 80 0.5040
  • DIAG 1 1 0.00470212 0.2133 0.6446
  • DIAG 1 1 0.00075280 0.0078
  • DIAG 1 1 0.00196054 0.0675 0.7953
  • DIAG 1 1 0.39391010 4 .0260
  • DIAG 1 1 0.20300 4 86 0.7516
  • Gender 1 1 0.00208025 0.0504 0.8226
  • DIAG 1 1 0.3139331 0.6930 0.4061
  • DIAG 1 1 0.02039153 0.2132 0.6447
  • DIAG 1 1 0.00477385 0.1678 0.6825
  • DIAG 1 1 0.0070432 0.1998 0.6554
  • DIAG 1 1 0.05169529 0.8901 0.3 4 65
  • Table 9 UPenn Plasma Alzheimer's disease vs Other Univariate. Age and Gender
  • DIAG 1 1 0.00309601 0.1352 0.7137

Abstract

The present invention includes novel markers useful for diagnosing Alzheimer's disease in an individual. The markers are also useful for qualifying Alzheimer's disease status in an individual. In particular, the markers can be detected an individual's plasma sample to classify the sample as Alzheimer's disease or non- Alzheimer's disease.

Description

TITLE OF THE INVENTION
Plasma Biomarkers for Diagnosis of Alzheimer's Disease
BACKGROUND OF THE INVENTION
Alzheimer's disease is a progressive neurodegenerative disorder estimated to affect 27 million people worldwide with numbers doubling every 20 years (Prince et al, 2009 Alzheimer's Disease International 1-92). Although symptoms of Alzheimer's disease manifest early as deficits in memory and other cognitive domains, pathological data show neuropathological features of Alzheimer's disease, including amyloid plaques and neurofibrillary tangles, occur well before the onset of dementia (Sawa et al, 2009 N Engl J Med. 360(22):2302-2309). Research focused upon cognitive performance in individuals who have progressed to dementia have further specified a pre-demented stage characterized by deficits in memory and executive function (Petersen et al, 2008 CNS Spectr. 13(l):45-53). Furthermore, pre-demented stages of Alzheimer's disease appear to be accompanied by a stereotypical biomarker signature supporting the notion that Alzheimer's disease can be diagnosed prior to the onset of dementia and that clinical studies focused upon dementia prevention are feasible (Craig-Schapiro et al, 2009 Neurobiol Dis. 35(2): 128-140).
Despite recent advances in the field, the diagnosis of Alzheimer's disease has not changed dramatically over the last 25 years. Currently, two major guidelines are utilized to make a probabilistic diagnosis of dementia of the Alzheimer's type (DAT). These include criteria specified in the Diagnostic and Statistical manual of Mental Disorders, fourth edition (DSM-IV-TR) and those delineated by the National Institute of Neurological Disorders and Stroke - Alzheimer's Disease and Related Disorders (NINCDS-ADRDA). DSM-IV criteria require the presence of both a memory disorder and impairment in at least one additional cognitive domain leading to disability in social function and/or day-day activities (APA Diagnostic and Statistical Manual of Mental Disorders (TV-TR), 4th ed text revised. Washington, DC; 2000). NINCDS-ADRDA are similar, but specify that onset is gradual and other systemic or neural diseases known to impair cognition have been excluded (McKhann et al., 1984 Neurology 34(7): 939-944). Most clinical studies in Alzheimer's disease utilize either NINCDS-ADRDA or DSM-IV criteria to enroll mild to moderate stage individuals. However, it is generally recognized that individuals with dementia are already in mid to late neuropathological stages of Alzheimer's disease. It remains unclear whether Alzheimer's disease-related neuropathology is reversible once dementia manifests. Indeed, recent studies have implied that amelioration of plaque load may occur following therapeutic treatment, but such clearance may not be associated with clinical benefit once the underlying pathology is significant. These types of reports raise concern around testing disease modifying therapies in mild-moderate Alzheimer's disease where neuropathology may already be significantly advanced, and suggest that the ability to maximize clinical benefit for disease modifying strategies may rely heavily upon early intervention, prior to significant and perhaps irreversible, neuropathological sequealae.
Although the diagnosis of Alzheimer's disease, prior to the onset of dementia remains challenging, a number of recent biomarker studies have suggested that symptomatic individuals at risk of progressing to dementia do exhibit a stereotypical biomarker phenotype. In brief, symptomatic, and in some cases non-symptomatic, individuals at risk of progressing to dementia exhibit decreased levels of CSF amyloid beta peptide 42 (Αβ42) and elevated levels of tau and phosphorylated tau (Mattsson et al, 2009 Jama 302(4):385-393; Shaw et al, 2009 Ann Neurol. 65(4):403-413; Sunderland et al, 2003 Jama 289(16):2094-2103; Visser et al, 2009 Lancet Neurol. 8(7):619-627). In addition, a number of imaging studies have suggested that symptomatic individuals at risk of progressing to dementia show increased hippocampal and brain atrophy, decreased glucose metabolism and increased amyloid PET labeling (Jack et al, 2009 Brain 132(Pt 5): 1355-1365; Landau et al, 2009 Neurobiol Aging. Aug 4; Leow et al, 2009
Neuroimage). The robustness of the data have led to position papers championing the use of disease specific biomarkers, such as amyloid PET labeling or CSF Αβ42 and tau, to aid in the identification of pre-dementia Alzheimer's disease (sometimes referred to as prodromal or early Alzheimer's disease) (Dubois et al, 2007 Lancet Neurol. 6(8):734- 746). As a result, clinical trials targeted at cognitive improvement and dementia prevention have begun to enroll individuals characterized as "early Alzheimer's disease" using a combination of memory deficits and CSF or PET amyloid markers.
Unfortunately, the use of CSF and imaging biomarkers as screening tools is proving to be quite challenging especially in countries where CSF and or imaging approaches may not be standard of care. As a result, the need for an intermediate, non-invasive, cost-effective screening tool is pressing.
While CSF and imaging biomarkers hold great promise as tools to enable early Alzheimer's disease drug development, they are problematic as diagnostic tools for the general practitioner. CSF procedures in many countries are considered highly invasive and PET imaging techniques are expensive and, in some countries, not widely available. Simple cognitive tests offer an alternative screening approach, but require time that is often not reimbursed by many health care systems. In the setting of a general practitioner where time constraints with individuals are an issue, cognitive tests are often not brief enough to accommodate a standard six-ten minute individual interview. A blood test would be a simple route for screening, and, if positive, could be used in combination with more extensive cognitive testing followed by referral to a specialist for more confirmatory CSF and imaging testing that would enable early diagnosis. A simple blood test would offer a tractable option towards screening individuals at risk. Although numerous studies have attempted to identify a blood based signature for Alzheimer's disease, success has been elusive. In 2007, Ray et al. (2007 Nat Med. 13(1 1): 1359-1362), reported on a multiplex blood based panel for identifying symptomatic individuals who progress to dementia. Many of the analytes described by Ray et al. (2007 Nat Med. 13(1 1): 1359-1362), are currently available on a commercially luminex xMAP multiplex panel.
There is an unmet need for simple biochemical tests that can detect Alzheimer's disease at an early stage, monitor progression of the disease, and
discriminate between Alzheimer's disease, normal individuals, non-Alzheimer's disease dementias and other neurological disorders. The present invention fills these needs by providing among other things, novel biomarkers and combinations of biomarkers useful for diagnosing Alzheimer's disease, as well as methods and kits for using the biomarkers to diagnose Alzheimer's disease.
SUMMARY OF THE INVENTION
The present invention provides a composition for detecting Alzheimer's
Disease in an individual with at least 80% sensitivity, comprising reagents for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual. Preferably, the biological sample is a plasma sample.
In one embodiment, the biomarkers for Alzheimer's disease comprise Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
In one embodiment, the biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309. In one embodiment, the biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
In one embodiment, the biomarkers include one or more biomarkers selected from Table 2.
In one embodiment, the reagent for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual is an antibody. In one embodiment, the reagent is on a solid support.
The invention provides a method of diagnosing Alzheimer's Disease in an individual, the method comprising analyzing a biological sample from an individual to determine the level(s) of one or more biomarkers for Alzheimer's Disease in the sample, wherein the one or more biomarkers are selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to diagnose whether the individual has Alzheimer's Disease.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to diagnose whether the individual has Alzheimer's Disease.
In one embodiment, the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
In one embodiment, the algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K- Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof. In one embodiment, the Alzheimer's Disease diagnosed is selected from the group consisting of late onset Alzheimer's disease, early onset Alzheimer's disease, familial Alzheimer's disease, and sporadic Alzheimer's disease.
In one embodiment, the biological sample is selected from the group consisting of whole blood, a blood component, CSF, urine, and any combination thereof.
The present invention provides a method for determining whether dementia in an individual is associated with AD, the method comprising analyzing a biological sample from an individual to determine the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin in the sample, and comparing the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP-2, and resistin in a corresponding reference sample to determine whether dementia in an individual is associated with AD.
In one embodiment, the dementia is associated with AD.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine the presence or status of dementia in the individual.
In one embodiment, the biological sample from the individual is analyzed to determine the level(s) of one or more biomarkers selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to determine the presence or status of Alzheimer's Disease or other types of dementia in the individual.
In one embodiment, the levels of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, resistin biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine the presence or status of Alzheimer's Disease or other types of dementia in the individual.
The present invention also includes a method for diagnosing Alzheimer's disease in an individual, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample from the individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive control reference values and optionally to negative control reference values, wherein levels of the biomarkers in the plasma sample of the individual resemble the positive control reference values and do not resemble the negative control reference values are an indication that the individual has Alzheimer's disease. In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the test individual in order to diagnose whether the individual has Alzheimer's Disease.
In one embodiment, the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
The present invention includes a method of determining risk of an individual developing Alzheimer's disease, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive and optionally to negative control values, wherein levels of the biomarkers in the plasma sample of the individual that resemble the positive control and do not resemble the negative control are an indication that the individual is at risk of developing Alzheimer's disease.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine risk of an individual of developing Alzheimer's disease.
In one embodiment, the level of biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine risk of an individual of developing Alzheimer's disease.
The invention includes a method for monitoring Alzheimer's disease in an individual, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual at a first time, and comparing the level of the biomarkers in the plasma sample of the individual at the first time to a level of the biomarkers in a plasma sample of the individual at the second time.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to monitor Alzheimer's disease in the individual.
In one embodiment, the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to monitor Alzheimer's disease in the individual.
The invention provides a method of assessing increased risk of developing Alzheimer's disease, the method comprising: obtaining a first sample of plasma from an individual blood at a first time, assessing the level of a biomarker for Alzheimer's disease in the first plasma sample to obtain a baseline level, obtaining a second sample of plasma from the individual at a second time to obtain a second level, assessing the level of the biomarker in the second plasma sample to obtain a second level, wherein if the second level is greater in the case of a biomarker that is over-expressed in AD or lower in the case of a biomarker that is under-expressed in AD, the individual is at an increased risk of developing Alzheimer's disease.
In one embodiment, the biomarker is one or more biomarkers selected from Table 2.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
In one embodiment, the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
The invention includes a method of assessing the likelihood that a pharmaceutical agent is efficacious in treating Alzheimer's disease in an individual, the method comprising: obtaining a first sample of plasma from an individual in the absence of a pharmaceutical agent, assessing the level of a biomarker for Alzheimer's disease in the first plasma sample to obtain a baseline level, administering the pharmaceutical agent to the individual, obtaining a second sample of plasma from the individual after administration of the pharmaceutical agent, assessing the level of the biomarker for Alzheimer's disease in the second plasma sample to obtain a treated level, wherein the likelihood that the pharmaceutical agent treats Alzheimer's disease is increased if the treated level is lower than the baseline level in the case of a biomarker that is over- expressed in AD and higher than the baseline level in the case of a biomarker that is under-expressed in AD.
In one embodiment, the biomarker is one or more biomarkers selected from Table 2.
In one embodiment, the method further comprises analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to assess the likelihood that the pharmaceutical agent is efficacious in treating
Alzheimer's disease in the individual. In one embodiment, the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess the likelihood that the pharmaceutical agent is efficacious in treating Alzheimer's disease in the individual.
The invention includes a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE provides a positive diagnosis of AD in the individual.
The invention includes a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides a positive diagnosis of AD in the individual.
In one embodiment, the method further comprises conducting one or more cognitive tests on the individual to confirm the positive diagnosis of AD.
In one embodiment, the method further comprises obtaining a CSF sample from the individual to confirm the positive diagnosis of AD.
The invention provides a method for screening to identify individuals at increased risk of developing AD, the method comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and comparing the levels of each of the at least five biomarkers to non-AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE provides an identification of an individual at increased risk of developing AD.
The invention provides a method for screening to identify individuals at increased risk of developing AD, the method comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and comparing the levels of each of the at least five biomarkers to non- AD controls, wherein a statistically significant increase in the levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides an identification of an individual at increased risk of developing AD.
The invention provides a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and determining whether, relative to non-AD controls, the levels of the at least five biomarkers provide a signature of a positive diagnosis of AD in the individual, wherein the signature comprises: a statistically significant increase in the levels of each of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in the level of ApoE.
The invention provides a method of diagnosing Alzheimer's Disease in an individual, comprising obtaining a plasma sample from the individual; assaying levels of at least five biomarkers in the plasma sample, wherein the at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and determining whether, relative to non-AD controls, the levels of the at least five biomarkers provide a signature of a positive diagnosis of AD in the individual, wherein the signature comprises: a statistically significant increase in the levels of each of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin.
The invention provides a kit for detecting Alzheimer's Disease in an individual with at least 80% sensitivity, comprising a reagent for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual.
In one embodiment, the reagent is capable of detecting biomarkers of
Alzheimer's disease in a plasma sample.
In one embodiment, the biomarkers of Alzheimer's disease comprise Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
In one embodiment, the biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309.
In one embodiment, the biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
In one embodiment, the biomarkers include one or more biomarkers selected from Table 2.
In one embodiment, the reagent is an antibody.
In one embodiment, the reagent is on a solid support.
In one embodiment, the kit further comprises an instruction manual.
BRIEF DESCRIPTION OF THE DRAWINGS
For the purpose of illustrating the invention, there are depicted in the drawings certain embodiments of the invention. However, the invention is not limited to the precise arrangements and instrumentalities of the embodiments depicted in the drawings.
Figure 1 is an image depicting Test set ROC curves for each model. The solid black dot indicates the default cutoff of 0.50 and the square indicates the alternate cutoff to get sensitivity closest to 0.80. ROC curves used 190 analytes and age, gender and ApoE4 allele and top 30 analytes from Partial Least Square Model (PLS).
Figure 2 is a series of graphs depicting expression of analytes in CSF Autopsy Confirmed Samples. Graphs represent means and error bars represent SEM.
Figure 3 is a graph depicting ROC curves using logistic regression of baseline (age, gender and ApoE genotype) vs Rules-Based Medicine (RBM) analytes alone versus baseline plus 24 RBM analytes. Thirty two total features in the final model: Age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), alphal microglobulin, Apolipoprotein E, angiopoietin-2, beta2 macroglobulin, BLC, Cortisol, E- selectin, Fatty acid binding protein, FAS, HGF, IGFBP-2, IL-10, NT proBNP,
Osteopontin, Pancreatic polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM, VEGF and von Willebrand Factor.
Figure 4A is an image demonstrating a correlation matrix analysis in the Alzheimer's disease samples exhibiting a high degree of correlation amongst the tested analytes. Figure 4B is a chart demonstrating high correlation of the analytes with each other and that there is high correlation with other analytes on the panel. For example, NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway. Without wishing to be bound by any particular theory, it is believed that analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance.
Figure 5 is an image demonstrating that plasma levels of ApoE, CRP and IL-15 differ dependent upon genetic ApoE allele status independent of diagnosis. Levels of ApoE4 are lowest in subjects with one or more E4 allele(s) and highest in subjects with one or more E2 allele(s).
Figure 6 is an image depicting graphical summary of the training and test set estimates for sensitivity and specificity
Figure 7 is an image depicting test set sensitivity and specificity profiles across various cutoffs for each model.
Figure 8 is an image depicting the overall distribution of PLS variables importance scores.
Figure 9 is an image depicting the correlation matrix for the top 15 predictors.
Figure 10 is an image depicting the training set data points for the top 15 predictors.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a method for diagnosing, monitoring and/or staging neurological disorders comprising the use of a multivariate and/or univariate approach to identify a neurological disorder in an individual.
The present invention relates generally to diagnostic methods and markers, prognostic methods and markers, and therapy evaluators for neurodegenerative disorders, such as Alzheimer's Disease. In one embodiment, the markers of the invention are useful for detecting early stage Alzheimer's disease.
In certain embodiments, the method comprises the step of obtaining a sample of plasma from the individual's blood, and assessing the level of markers of Alzheimer's Disease in the plasma sample. Thus, the present invention relates to markers of Alzheimer's Disease, methods for diagnosis of Alzheimer's Disease, methods of determining predisposition to Alzheimer's Disease, methods of monitoring
progression/regression of Alzheimer's Disease, methods of assessing efficacy of compositions for treating Alzheimer's Disease, methods of screening compositions for activity in modulating markers of Alzheimer's Disease, methods of treating Alzheimer's Disease, as well as other methods based on markers of Alzheimer's Disease.
In certain embodiments, the invention further provides methods for permitting refinement of disease diagnosis, disease risk prediction, and clinical management of individuals associated with a neurodegenerative disorder. The
Alzheimer's disease markers of the invention represent a plasma based panel for assessing Alzheimer's disease that can be used for determining the disease state or disease risk. The detection of the selective markers of the invention in individuals, or samples obtained therefrom permits refinement of disease diagnosis, disease risk prediction, and clinical management of individuals being treated with agents that are associated with Alzheimer's disease.
In one embodiment, the Alzheimer's disease markers of the invention include one or more of the markers shown in Table 2.
In one embodiment of the invention, age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in identifying an individual for Alzheimer's disease.
In one embodiment of the invention, markers of AD includes at least
Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
In one embodiment of the invention, markers of AD includes at least resistin, e3/e3, pancreatic polypeptide, e3/e4, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, and 1-309.
In one embodiment of the invention, markers of AD includes at least
Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein,
Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor. In one embodiment, the plasma based panel for assessing Alzheimer's disease comprises one or more of the following markers: Alpha-1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E- Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2,
Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases- 1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor. In some embodiments, age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
The invention includes a method comprising obtaining a sample from an individual, and assessing the level of one or more of Alpha-1 Microglobulin,
Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, and Von Willebrand Factor in the sample. However, the invention should not be limited to only these markers disclosed herein (e.g., Table 2) because a skilled artisan when armed with the present disclosure would be able identify additional markers that can be used as indicators for Alzheimer's disease. For example, as discussed in the Examples, a test sample and a control sample can be subjected to any commercially available panel comprising a plurality of markers and analyzed according to the statistic models disclosed herein to identify markers associated with AD.
Moreover, the disclosure presented here demonstrates a high degree of correlation amongst certain markers for identifying AD in an individual. A skilled artisan when armed with the present disclosure, particularly Figures 4A and 4B, would recognize that analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance. For example, NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway. Thus, markers that are associated with an AD specific pathophysiological pathway can be interchangeable. Accordingly, correlation amongst the markers of the invention provides means to identify other related markers associated with the specific pathophysiological pathway as being indicators for Alzheimer's disease.
Any number of biomarkers may be used in the methods disclosed herein. That is, the disclosed methods may include the determination of the level(s) of one biomarker, two or more biomarkers, three or more biomarkers, four or more biomarkers, five or more biomarkers, six or more biomarkers, seven or more biomarkers, eight or more biomarkers, nine or more biomarkers, ten or more biomarkers, fifteen or more biomarkers, etc., including a combination of all of the biomarkers in each or all of Tables 2, 3, 7, 8, 9, 10, 1 1, 12 or any fraction thereof. In another aspect, the biomarkers are used in combination with other factors that predict for AD. Such factors include but are not limited to age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4).
In one embodiment, an immunoassay is used for the assessment of a marker level. In another embodiment, a luminex technology multiplex immunoassay is used to assess the marker level.
In another embodiment, a method of diagnosing Alzheimer's disease in an individual comprises the steps of obtaining a first sample of plasma from the individual at a first time; assessing the level of Alzheimer's disease markers using plasma based panel of the invention in the first plasma sample to obtain a baseline level; obtaining a second sample of plasma from the individual at a second time and assessing the level of
Alzheimer's disease markers in the second plasma sample to obtain a second level. If the second level is significantly altered compared to the baseline level, the individual is at an increased risk of developing or having Alzheimer's disease. In one embodiment, the second level is also compared to a reference population of an individual's without Alzheimer's disease; if the second level is significantly altered compared to the level derived from a reference population, the individual is at an increased risk of developing or having Alzheimer's disease.
In still further embodiments, the invention provides methods of monitoring a plasma panel of particular markers of Alzheimer's disease to evaluate the progress of a therapeutic treatment of Alzheimer's disease.
In another embodiment, the invention provides methods for selecting a patient that is most likely to respond to treatment.
The invention also provides methods for screening an individual to determine if the individual is at increased risk of having Alzheimer's disease. Individuals found to be at increased risk can be given appropriate therapy and monitored using the methods of the invention.
Other methods and kits useful in practicing the methods of the invention are provided herein.
Definitions
As used herein, each of the following terms has the meaning associated with it in this section.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
"About" as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or ±10%, more preferably ±5%, even more preferably ±1%, and still more preferably ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
The term "abnormal" when used in the context of organisms, tissues, cells or components thereof, refers to those organisms, tissues, cells or components thereof that differ in at least one observable or detectable characteristic (e.g., age, treatment, time of day, etc.) from those organisms, tissues, cells or components thereof that display the "normal" (expected) respective characteristic. Characteristics which are normal or expected for one cell or tissue type, might be abnormal for a different cell or tissue type.
The term "antibody," as used herein, refers to an immunoglobulin molecule which is able to specifically bind to a specific epitope on an antigen.
Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins. The antibodies in the present invention may exist in a variety of forms including, for example, polyclonal antibodies, monoclonal antibodies, intracellular antibodies
("intrabodies"), Fv, Fab and F(ab)2, as well as single chain antibodies (scFv), heavy chain antibodies, such as camelid antibodies, and humanized antibodies (Harlow et al, 1999, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al, 1989, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York; Houston et al, 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al, 1988, Science 242:423-426). By the term "synthetic antibody" as used herein, is meant an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage as described herein. The term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
As used herein, the term "heavy chain antibody" or "heavy chain antibodies" comprises immunoglobulin molecules derived from camelid species, either by immunization with a peptide and subsequent isolation of sera, or by the cloning and expression of nucleic acid sequences encoding such antibodies. The term "heavy chain antibody" or "heavy chain antibodies" further encompasses immunoglobulin molecules isolated from an animal with heavy chain disease, or prepared by the cloning and expression of VH (variable heavy chain immunoglobulin) genes from an animal.
As used herein, an "immunoassay" refers to any binding assay that uses an antibody capable of binding specifically to a target molecule to detect and quantify the target molecule.
By the term "specifically binds," as used herein with respect to an antibody, is meant an antibody which recognizes an specific antigen, but does not substantially recognize or bind other molecules in a sample. For example, an antibody that specifically binds to an antigen from one species may also bind to that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific. In another example, an antibody that specifically binds to an antigen may also bind to different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific.
In some instances, the terms "specific binding" or "specifically binding", can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A", the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A" and the antibody, will reduce the amount of labeled A bound to the antibody. As used herein, "biomarker" in the context of the present invention encompasses, without limitation, proteins, nucleic acids, and metabolites, together with their polymorphisms, mutations, variants, modifications, subunits, fragments, protein- ligand complexes, and degradation products, protein-ligand complexes, elements, related metabolites, and other analytes or sample-derived measures. Biomarkers can also include mutated proteins or mutated nucleic acids. Biomarkers also encompass non-blood borne factors or non-analyte physiological markers of health status, such as clinical parameters, as well as traditional laboratory risk factors. Biomarkers also include any calculated indices created mathematically or combinations of any one or more of the foregoing measurements, including temporal trends and differences.
As used herein, the term "data" in relation to one or more biomarkers, or the term "biomarker data" generally refers to data reflective of the absolute and/or relative abundance (level) of a product of a biomarker in a sample. As used herein, the term "dataset" in relation to one or more biomarkers refers to a set of data representing levels of each of one or more biomarker products of a panel of biomarkers in a reference population of subjects. A dataset can be used to generate a formula/classifier of the invention. According to one embodiment the dataset need not comprise data for each biomarker product of the panel for each individual of the reference population. For example, the "dataset" when used in the context of a dataset to be applied to a formula can refer to data representing levels of products of each biomarker for each individual in one or more reference populations, but as would be understood can also refer to data representing levels of products of each biomarker for 99%, 95%, 90%, 85%, 80%, 75%, 70% or less of the individuals in each of said one or more reference populations and can still be useful for purposes of applying to a formula.
"Differentially increased expression" or "up regulation" refers to biomarker product levels which are at least 10% or more, for example, 20%, 30%, 40%, or 50%, 60%, 70%, 80%, 90% higher or more, and/or 1.1 fold, 1.2 fold, 1.4 fold, 1.6 fold, 1.8 fold higher or more, than a control.
"Differentially decreased expression" or "down regulation" refers to biomarker product levels which are at least 10% or more, for example, 20%, 30%, 40%, or 50%, 60%, 70%, 80%, 90% lower or less, and/or 0.9 fold, 0.8 fold, 0.6 fold, 0.4 fold, 0.2 fold, 0.1 fold or less lower than a control.
A "formula," "algorithm," or "model" is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous or categorical inputs (herein called "parameters") and calculates an output value, sometimes referred to as an "index" or "index value." Non-limiting examples of "formulas" include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations. Of particular use in combining Alzheimer's disease markers and other biomarkers are linear and non-linear equations and statistical classification analyses to determine the relationship between levels of Alzheimer's disease markers detected in a subject sample. In panel and combination construction, of particular interest are structural and synactic statistical classification algorithms, and methods of risk index construction, utilizing pattern recognition features, including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Linear Discriminant Analysis (LDA), Support Vector Machines (SVM), Random Forest (RF), Partial Least Squares, Sparse Partial Least Squares, Flexible Discriminant Analysis, Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Nearest Shrunken Centroids (SC)", stepwise model selection procedures, Kth-Nearest Neighbor, Boosting or Boosted Tree, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others. Other techniques may be used in survival and time to event hazard analysis, including Cox, Weibull, Kaplan-Meier and Greenwood models well known to those of skill in the art.
An "analyte", as used herein refers to any substance or chemical constituent that is undergoing analysis. For example, an "analyte" can refer to any atom and/or molecule; including their complexes and fragment ions. The term may refer to a single component or a set of components. In the case of biological
molecules/macromolecules, such analytes include but are not limited to: polypeptides, polynucleotides, proteins, peptides, antibodies, DNA, RNA, carbohydrates, steroids, and lipids, and any detectable moiety thereof, e.g. immunologically detectable fragments.
A "disease" is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate. A "disorder" in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
As used herein, the terms "Alzheimer's disease" and "Alzheimer's disease" refer to a neurodegenerative disorder and encompass familial Alzheimer's disease and sporadic Alzheimer's disease. The term "familial Alzheimer's disease" refers to
Alzheimer's disease associated with genetic factors (i.e., inheritance is demonstrated) while "sporadic Alzheimer's disease" refers to Alzheimer's disease that is not associated with prior family history of the disease. Symptoms indicative of Alzheimer's disease in human subjects typically include, but are not limited to, mild to severe dementia, progressive impairment of memory (ranging from mild forgetfulness to disorientation and severe memory loss), poor visual spatial skills, personality, changes, poor impulse control, poor judgment, distrust of others, increased stubbornness, restlessness, poor planning ability, poor decision making, and social withdrawal. In severe cases, patients lose the ability to use language and communicate, and require assistance in personal hygiene, eating and dressing, and are eventually bedridden. Hallmark pathologies within brain tissue include extracellular neuritic amyloid plaques, neurofibrillary tangles, neurofibrillary degeneration, granulovascular neuronal degeneration, synaptic loss, and extensive neuronal cell death.
"Increased risk of developing Alzheimer's disease" is used herein to refer to an increase in the likelihood or possibility of developing Alzheimer's disease. This risk can be assessed relative to an individual's own risk, or with respect to a reference population that does not have clinical evidence of Alzheimer's disease. The reference population may be representative of the individual with regard to approximate age, age group and/or gender.
"Delaying development of Alzheimer's disease" as used herein refers to a prolonging of the time to the development of Alzheimer's disease and/or delay in the rate of increased extent of Alzheimer's disease.
"Alleviating Alzheimer's disease" as used herein refers to a decrease in the severity of Alzheimer's disease.
As used herein, the terms "Alzheimer's patient", "Alzheimer's disease patient", and "individual diagnosed with Alzheimer's disease" all refer to an individual who has been diagnosed with Alzheimer's disease or has been given a probable diagnosis of Alzheimer's Disease.
An "individual with mild Alzheimer's disease" is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable Alzheimer's disease. In some instances, an "individual with mild Alzheimer's disease" has either been assessed with the Mini-Mental State Examination (MMSE) (referenced in Folstein et al, J. Psychiatr. Res 1975; 12: 1289-198) and scored 22-27 or would achieve a score of 22-27 upon MMSE testing.
An "individual with moderate Alzheimer's disease" is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable Alzheimer's disease. In some instances, an "individual with moderate Alzheimer's disease" has either been assessed with the MMSE and scored 16-21 or would achieve a score of 16-21 upon MMSE testing.
An "individual with severe Alzheimer's disease" is an individual who has been diagnosed with Alzheimer's disease or has been given a diagnosis of probable
Alzheimer's disease. In some instances, an "individual with severe Alzheimer's disease" has either been assessed with the MMSE and scored 12-15 or would achieve a score of 12-15 upon MMSE testing.
The "level" of one or more biomarkers means the absolute or relative amount or concentration of the biomarker in the sample.
"Measuring" or "measurement," or alternatively "detecting" or
"detection," means assessing the presence, absence, quantity or amount (which can be an effective amount) of either a given substance within a clinical or subject-derived sample, including the derivation of qualitative or quantitative concentration levels of such substances, or otherwise evaluating the values or categorization of a subject's clinical parameters.
A "reference level" of a biomarker means a level of the biomarker that is indicative of a particular disease state, phenotype, or lack thereof, as well as combinations of disease states, phenotypes, or lack thereof. A "positive" reference level of a biomarker means a level that is indicative of a particular disease state or phenotype. A "negative" reference level of a biomarker means a level that is indicative of a lack of a particular disease state or phenotype. For example, an "Alzheimer's Disease-positive reference level" of a biomarker means a level of a biomarker that is indicative of a positive diagnosis of Alzheimer's Disease in a subject, and an "Alzheimer's Disease-negative reference level" of a biomarker means a level of a biomarker that is indicative of a negative diagnosis of Alzheimer's Disease in a subject. As another example, an
"Alzheimer's-Disease-progression-positive reference level" of a biomarker means a level of a biomarker that is indicative of progression of Alzheimer's Disease in a subject, and an "Alzheimer's-Disease-regression-positive reference level" of a biomarker means a level of a biomarker that is indicative of regression of the Alzheimer's Disease. A "reference level" of a biomarker may be an absolute or relative amount or concentration of the biomarker, a presence or absence of the biomarker, a range of amount or concentration of the biomarker, a minimum and/or maximum amount or concentration of the biomarker, a mean amount or concentration of the biomarker, and/or a median amount or concentration of the biomarker; and, in addition, "reference levels" of combinations of biomarkers may also be ratios of absolute or relative amounts or concentrations of two or more biomarkers with respect to each other. Appropriate positive and negative reference levels of biomarkers for a particular disease state, phenotype, or lack thereof may be determined by measuring levels of desired biomarkers in one or more appropriate subjects, and such reference levels may be tailored to specific populations of subjects (e.g., a reference level may be age-matched so that comparisons may be made between biomarker levels in samples from subjects of a certain age and reference levels for a particular disease state, phenotype, or lack thereof in a certain age group). Such reference levels may also be tailored to specific techniques that are used to measure levels of biomarkers in biological samples, where the levels of biomarkers may differ based on the specific technique that is used. Such reference levels may also be tailored to specific statistical models used to analyze levels of biomarkers in biological samples, where the levels of biomarkers may differ based on the specific statistical model that is used.
As used herein, a "detector molecule" is a molecule that may be used to detect a compound of interest. Non-limiting examples of a detector molecule are molecules that bind specifically to a compound of interest, such as, but not limited to, an antibody, a cognate receptor, and a small molecule.
An "effective amount" or "therapeutically effective amount" of a compound is that amount of compound which is sufficient to provide a beneficial effect to the individual to which the compound is administered. An "effective amount" of a delivery vehicle is that amount sufficient to effectively bind or deliver a compound.
As used herein, an "instructional material" includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of a compound, composition, vector, or delivery system of the invention in the kit for effecting alleviation of the various diseases or disorders recited herein. Optionally, or alternately, the instructional material can describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a individual. The instructional material of the kit of the invention can, for example, be affixed to a container which contains the identified compound, composition, vector, or delivery system of the invention or be shipped together with a container which contains the identified compound, composition, vector, or delivery system. Alternatively, the instructional material can be shipped separately from the container with the intention that the instructional material and the compound be used cooperatively by the recipient.
The term "microarray" refers broadly to both "DNA microarrays" and "DNA chip(s)," and encompasses all art-recognized solid supports, and all art-recognized methods for affixing nucleic acid molecules thereto or for synthesis of nucleic acids thereon.
The terms "patient," "subject," "individual," and the like are used interchangeably herein, and refer to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein. In certain non-limiting embodiments, the patient, subject or individual is a human.
The term "protein" typically refers to large polypeptides.
"Sample" or "biological sample" as used herein means a biological material isolated from an individual. The biological sample may contain any biological material suitable for detecting the desired biomarkers, and may comprise cellular and/or non-cellular material obtained from the individual.
A "therapeutic" treatment is a treatment administered to an individual who exhibits signs of pathology, for the purpose of diminishing or eliminating those signs.
The term to "treat," as used herein, means reducing the frequency with which symptoms are experienced by an individual or subject or administering an agent or compound to reduce the frequency with which symptoms are experienced. In some instances, "treat" and "treating" are not limited to the case where the subject (e.g. patient) is cured and the disease is eradicated. Rather, the present invention also contemplates treatment that merely reduces symptoms, improves (to some degree) and/or delays disease progression. The term "treatment" also refers to the alleviation, amelioration, and/or stabilization of symptoms, as well as delay in progression of symptoms of a particular disorder. For example, "treatment" of Alzheimer's disease includes any one or more of: elimination of one or more symptoms of Alzheimer's disease, reduction of one or more symptoms of Alzheimer's disease, stabilization of the symptoms of Alzheimer's disease (e.g., failure to progress to more advanced stages of Alzheimer's disease), and delay in progression (i.e., worsening) of one or more symptoms of Alzheimer's disease.
As used herein, "treating a disease or disorder" means reducing the frequency with which a symptom of the disease or disorder is experienced by an individual. Disease and disorder are used interchangeably herein.
Ranges: throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range and, when appropriate, partial integers of the numerical values within ranges. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range. Description
Alzheimer's disease is a progressive neurodegenerative disorder characterized by memory and cognitive impairments and other non-cognitive behavioral symptoms. Age is the strongest risk factor, wherein almost 50% of people over the age of 85 are affected. Early-onset Alzheimer's disease (EOAD) is associated with genetic mutations in amyloid precursor protein (APP), presenilin 1 (PSE 1) and presenilin 2
(PSEN2). However, sporadic or late-onset Alzheimer's disease (LOAD) is multi-factorial and genetically more complex. In addition, genetic factors may account for as much as 80% of the disease risk associated with LOAD (Gatz et al. (2006) Arch. Gen. Psychiatry 63(2): 168-174). While monogenic mutations cause EOAD, the only extensively validated susceptibility gene for LOAD is the apolipoprotein E (ΑΡΟΕ-ε4) allele
(Saunders et al. (1993) Neurology 43(8): 1467-1472 and Farrer et al. (1997) JAMA 278(16): 1349-1356). But alleles of the APOE gene do not account for all of the genetic load responsible for LOAD predisposition. Currently, Alzheimer's can be identified through the use of cognitive tests combined with cerebrospinal fluid (CSF) and imaging biomarkers. However, such tests are often costly and invasive and are difficult to implement as screening tools to identify patient s suitable for clinical trial study.
The present invention provides novel Alzheimer's disease biomarkers present in a biological sample of an individual. Preferably, the biological sample is plasma. The level of these biomarkers allow a more accurate diagnosis or prognosis of Alzheimer's disease in individuals that are at risk for Alzheimer's disease, that exhibit no clinical signs of Alzheimer's disease, or that exhibit minor clinical signs of Alzheimer's disease. Furthermore, the measurement of the biomarkers of the invention allows the monitoring of Alzheimer's disease, such that a comparison of biomarker levels facilitates an evaluation of disease progression in individuals that have been diagnosed with Alzheimer's disease, or that do not yet exhibit any clinical signs of Alzheimer's disease. Moreover, the Alzheimer's disease biomarkers of the invention may be used in concert with known Alzheimer's disease biomarkers such that a more accurate diagnosis or prognosis of Alzheimer's disease may be made.
In one embodiment, the invention provides a screening tool to detect Alzheimer's disease in an individual. Preferably, the screening tool includes a examining a multiplexed plasma panel to differentiate Alzheimer's disease from age-matched controls. In some instances, the screening tool encompasses a multivariate based approach combined a statistical model to predict Alzheimer's disease in an individual. For example, the multivariate approach useful in the invention encompasses the use of a multiplexed plasma panel in combination with age, gender and ApoE genetic status (6 co- variates e2/e2; e2/e3; e2/e4; e3/e4; e3/e3; e4/e4) for determining Alzheimer's disease in an individual. In some instances, the screening tool of the invention can differentiate Alzheimer's disease from age-matched controls with high sensitivity suitable for preliminary screening of individuals who might be eligible for clinical trial enrollment in early Alzheimer's disease studies. The disclosure presented herein also includes additional markers associated with increased or decreased risk of Alzheimer's disease as shown in Table 2.
The present invention relates partly on the discovery of biomarkers that can differentiate Alzheimer's disease from controls and identify individuals at risk of progressing to dementia. A two tiered strategy was employed to find the optimal discriminatory panel of biomarkers. First, multivariate approaches were utilized to identify top plasma analytes that contributed to maximal differentiation using a training/test set approach. Second, plasma analytes were compared to expression in CSF from individuals with diagnosis confirmed by autopsy analysis. A top list of analytes was identified based upon 1) performance in multivariate models, 2) expression in CSF from autopsy confirmed individuals and 3) upon linkage to disease by pathway analysis. A final analyte panel of analytes, combined with age, gender and ApoE allele status, was then subjected to logistic regression analysis to determine performance in differentiating Alzheimer's disease from controls. An objective was to determine whether a plasma based signature with a sensitivity of at least 80% could be achieved. In some instances, the objective was to determine whether a plasma based signature with a sensitivity of at least 80% and a specificity of at least 80% could be achieved. A test with reasonable (at least 80%) sensitivity and specificity (Consensus report of the Working Group on:
"Molecular and Biochemical Markers of Alzheimer's Disease". The Ronald and Nancy Reagan Research Institute of the Alzheimer's Association and the National Institute on Aging Working Group. Neurobiol Aging. Mar-Apr 1998; 19(2): 109-116) could be useful as a screening tool to identify individuals at risk who would then be eligible for more confirmatory cognitive, CSF and imaging clinical trial enrollment testing.
Biomarkers to Detect Alzheimer's Disease
A biomarker is an organic biomolecule which is differentially present in a sample taken from an individual of one phenotypic status (e.g., having a disease) as compared with an individual of another phenotypic status (e.g., not having the disease).
A biomarker is differentially present between the two individuals if the mean or median expression level of the biomarker in the different individuals is calculated to be statistically significant. Biomarkers, alone or in combination, provide measures of relative risk that an individual belongs to one phenotypic status or another. Therefore, they are useful as markers for diagnosis of disease, therapeutic effectiveness of a drug, and drug toxicity.
Accordingly, the invention provides methods for identifying one or more biomarkers which can be used to aid in the diagnosis, to diagnose, detect, predict neurological diseases such as neurodegenerative disorders. The methods of the invention are carried out by obtaining a set of measured values for a plurality of biomarkers from a biological sample derived from a test individual, obtaining a set of measured values for a plurality of biomarkers from a biological sample derived from a control individual, comparing the measured values for each biomarker between the test and control sample, and identifying biomarkers which are significantly different between the test value and the control value also referred to as a reference value.
The process of comparing a measured value and a reference value can be carried out in any convenient manner appropriate to the type of measured value and reference value for the AD biomarker at issue. For example, "measuring" can be performed using quantitative or qualitative measurement techniques, and the mode of comparing a measured value and a reference value can vary depending on the
measurement technology employed. For example, when a qualitative calorimetric assay is used to measure AD biomarker levels, the levels may be compared by visually comparing the intensity of the colored reaction product, or by comparing data from densitometric or spectrometric measurements of the colored reaction product (e.g., comparing numerical data or graphical data, such as bar charts, derived from the measuring device). However, it is expected that the measured values used in the methods of the invention will most commonly be quantitative values (e.g., quantitative
measurements of concentration. In other examples, measured values are qualitative. As with qualitative measurements, the comparison can be made by inspecting the numerical data, by inspecting representations of the data (e.g., inspecting graphical representations such as bar or line graphs).
A measured value is generally considered to be substantially equal to or greater than a reference value if it is at least about 95% of the value of the reference. A measured value is considered less than a reference value if the measured value is less than about 95% of the reference value. A measured value is considered more than a reference value if the measured value is at least more than about 5% greater than the reference value.
The process of comparing may be manual (such as visual inspection by the practitioner of the method) or it may be automated. For example, an assay device (such as a luminometer for measuring chemiluminescent signals) may include circuitry and software enabling it to compare a measured value with a reference value for an AD biomarker. Alternately, a separate device (e.g., a digital computer) may be used to compare the measured value(s) and the reference value(s). Automated devices for comparison may include stored reference values for the AD biomarker(s) being measured, or they may compare the measured value(s) with reference values that are derived from contemporaneously measured reference samples. In some embodiments, the methods of the invention utilize "simple" or "binary" comparison between the measured level(s) and the reference level(s) (e.g., the comparison between a measured level and a reference level determines whether the measured level is higher or lower than the reference level). For AD biomarkers, a comparison showing that the measured value for the biomarker is altered from the reference value indicates or suggests a diagnosis of AD.
By way of a non-limiting example, the process of comparing the measured values to identify indicators for disease state can be carried out using multiple marker analysis approach including but not limited to Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes,
Partial Least Squares (PLS), Random Forests, Shrunken Centroids, Sparse Partial Least Squares and Support Vector Machines approaches.
One aspect of the present invention provides biomarkers to detect Alzheimer's disease. A biomarker is typically a protein, found in a bodily fluid, whose level varies with disease state and may be readily quantified. The quantified level may then be compared to a known value. The comparison may be used for several different purposes, including but not limited to, diagnosis of Alzheimer's disease, prognosis of Alzheimer's disease, and monitoring treatment of Alzheimer's disease.
Through screening performed as detailed in the examples provided elsewhere herein, several biomarkers have been identified as associated with Alzheimer's disease. Examples of biomarkers identified as being indicators of Alzheimer's disease include, but is not limited, to Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
Thrombomodulin, TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, and any combination thereof. In some instances, age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
In one aspect, the invention provides compositions for use in the methods of the invention. In one embodiment, the composition comprises a reagent that detects and/or quantitates an Alzheimer Disease biomarker. In some embodiments, the composition comprises a panel of reagents, each of which detects and/or quantitates a different Alzheimer Disease biomarker. In some embodiments, the Alzheimer Disease biomarker is an Alzheimer Disease biomarker shown in Table 2. In some embodiments, the composition comprises two or more reagents, each of which detects and/or quantitates a different one of two or more, three or more, four or more or five or more Alzheimer Disease biomarkers. In some embodiments, the compositions comprises a panel of reagents each of which detects and/or quantitates a different one of 24 Alzheimer Disease biomarkers shown in Table 2. In some embodiments, the composition comprises a panel of reagents for the detection and/or quantification of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin. In some embodiments, the composition comprises a panel of reagents for the detection and/or quantification of resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB,
MMP 1, and 1-309. In some embodiments, the composition comprises a panel of reagents for the detection and/or quantification of Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
In one embodiment, the combination of biomarkers associated with assessing Alzheimer's disease is collectively presented on a detectable medium referred to as a panel or plasma based panel. Each of the biomarkers identified above may be used in concert with other biomarkers. For instance, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 10 or more, 15 or more, 20 or more, 30 or more, 40 or more, (and any integer value in between) Alzheimer's disease biomarkers may be used in concert with other, and with other known or to be known biomarkers for
Alzheimer's disease.
In some embodiments, the composition is an array of two or more detection reagents, e.g., antibodies and/or oligonucleotides each of which binds to a different one of two Alzheimer Disease biomarker proteins or nucleic acids, respectively. In a preferred embodiment, the Alzheimer Disease biomarkers are proteins and the array contains antibodies sufficient to measure a statistically significant alteration in Alzheimer Disease biomarker expression compared to a reference value. In some embodiments, the invention provides a kit with at least two detection reagents, each of which binds to a different one of two Alzheimer Disease biomarker proteins, metabolites, or other analytes.
In some embodiments, a. detection reagent is immobilized on a solid matrix such as a porous strip or bead to form at least one Alzheimer Disease biomarker detection site,
Provided herein are methods for assessing cognitive function, assessing cognitive impairment, diagnosing or aiding diagnosis of cognitive impairment by obtaining measured levels of one or more AD diagnosis biomarkers in a biological sample from an individual, such as for example, a plasma sample from an individual, and comparing those measured levels to reference levels.
The levels of Alzheimer's disease biomarkers of the invention may be assessed in several different biological samples, for example bodily fluids. Non-limiting examples of bodily fluid include whole blood, plasma, serum, bile, lymph, pleural fluid, semen, saliva, sweat, urine, and CSF. In one embodiment, the bodily fluid is selected from the group of whole blood, plasma, and serum. In another embodiment, the bodily fluid is whole blood. In yet another embodiment, the bodily fluid is plasma. In still yet another embodiment, the bodily fluid is serum.
The bodily fluid is obtained from the individual using conventional methods in the art. For instance, one skilled in the art knows how to draw blood and how to process it in order to obtain serum and/or plasma for use in the method. Generally speaking, the method preferably maintains the integrity of the biomarkers of the invention such that it can be accurately quantified in the bodily fluid. Methods for collecting blood or fractions thereof are well known in the art. For example, see US Patent No. 5,286,262, which is hereby incorporated by reference in its entirety.
A bodily fluid may be obtained from any mammal known to suffer from Alzheimer's disease or that can be used as a disease model for Alzheimer's disease. In one embodiment, the mammal is a rodent. Examples of rodents include mice, rats, and guinea pigs. In another embodiment, the mammal is a primate. Examples of primates include monkeys, apes, and humans. In an exemplary embodiment, the mammal is a human. In some embodiments, the individual has no clinical signs of Alzheimer's disease. In other embodiments, the individual has mild clinical signs of Alzheimer's disease. In yet other embodiments, the individual may be at risk for Alzheimer's disease. In still other embodiments, the individual has been diagnosed with Alzheimer's disease. Assessment of biomarker levels may encompass assessment of the level of protein concentration or the level of enzymatic activity of the biomarker, wherever applies. In either case, the level is quantified such that a value, an average value, or a range of values is determined. In one embodiment, the level of protein concentration of the Alzheimer's disease biomarker is quantified.
There are numerous known methods and kits for measuring the amount or concentration of a protein in a sample, including as non-limiting examples, ELISA, western blot, absorption measurement, colorimethc determination, Lowry assay, Bicinchoninic acid assay, or a Bradford assay. Commercial kits include ProteoQwest™ Colohmetric Western Blotting Kits (Sigma-Aldrich, Co.), QuantiPro™ bicinchoninic acid (BCA) Protein Assay Kit (Sigma-Aldrich, Co.), FluoroProfile™ Protein Quantification Kit (Sigma-Aldrich, Co.), the Coomassie Plus - The Better Bradford Assay (Pierce Biotechnology, Inc.), and the Modified Lowry Protein Assay Kit (Pierce Biotechnology, Inc.). In certain embodiments, the protein concentration is measured using a luminex based multiplex immunoassay panel. However, the invention should not be limited to any particular assay for assessing the level of a biomarker of the invention. That is, any currently known assay used to detect protein levels and assays to be discovered in the future can be used to detect the biomarkers of the invention.
Methods of quantitatively assessing the level of a protein in a biological sample such as plasma are well known in the art. In some embodiments, assessing the level of a protein involves the use of a detector molecule for the biomarker. Detector molecules can be obtained from commercial vendors or can be prepared using conventional methods available in the art. Exemplary detector molecules include, but are not limited to, an antibody that binds specifically to the biomarker, a naturally-occurring cognate receptor, or functional domain thereof, for the biomarker, and a small molecule that binds specifically to the biomarker.
In a preferred embodiment, the level of a biomarker is assessed using an antibody. Thus, non-limiting exemplary methods for assessing the level of a biomarker in a biological sample include various immunoassays, for example, immunohistochemistry assays, immunocytochemistry assays, ELISA, capture ELISA, sandwich assays, enzyme immunoassay, radioimmunoassay, fluorescent immunoassay, and the like, all of which are known to those of skill in the art. See e.g. Harlow et ah, 1988, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York; Harlow et ah, 1999, Using
Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY. The generation of polyclonal antibodies is accomplished by inoculating the desired animal with an antigen and isolating antibodies which specifically bind the antigen therefrom.
Monoclonal antibodies directed against one of the biomarkers identified herein may be prepared using any well known monoclonal antibody preparation procedures, such as those described, for example, in Harlow et al. (1988, In: Antibodies, A Laboratory Manual, Cold Spring Harbor, NY) and in Tuszynski et al. (1988, Blood, 72: 109-1 15). Human monoclonal antibodies may be prepared by the method described in U.S. patent publication 2003/0224490. Monoclonal antibodies directed against a biomarker such as EGF are generated from mice immunized with the biomarker using standard procedures as referenced herein.
For use in preparing an antibody, a biomarker may be purified from a biological source that endogenously comprises the biomarker, or from a biological source recombinantly-engineered to produce or over-produce the biomarker, using conventional methods known in the art. Exemplary nucleic acid and protein sequences for the eleven biomarkers described herein are readily available in public sequence databases, such as National Library of Medicine's genetic sequence database GenBank® (Benson et al, 2008, Nucleic Acids Research, 36(Database issue):D25-30). Preferably, antibodies are generated against the human homologs of each of the eleven biomarkers for practicing the methods using a biological sample from a human individual diagnosed with Parkinson's Disease.
Nucleic acid encoding the monoclonal antibody obtained using the procedures described herein may be cloned and sequenced using technology which is available in the art, and is described, for example, in Wright et al. (1992, Critical Rev. Immunol. 12(3,4): 125-168) and the references cited therein. Further, the antibody useful in the practice of the invention may be "humanized" using the technology described in Wright et al, (supra) and in the references cited therein, and in Gu et al. (1997,
Thrombosis and Hematocyst 77(4):755-759).
To generate a phage antibody library, a cDNA library is first obtained from mRNA which is isolated from cells, e.g., the hybridoma, which express the desired protein to be expressed on the phage surface, e.g., the desired antibody. cDNA copies of the mRNA are produced using reverse transcriptase. cDNA which specifies
immunoglobulin fragments are obtained by PCR and the resulting DNA is cloned into a suitable bacteriophage vector to generate a bacteriophage DNA library comprising DNA specifying immunoglobulin genes. The procedures for making a bacteriophage library comprising heterologous DNA are well known in the art and are described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY).
Bacteriophage which encode the desired antibody may be engineered such that the protein is displayed on the surface thereof in such a manner that it is available for binding to the antigen against which the antibody is directed. Thus, when bacteriophage which express a specific antibody are incubated in the presence of the antigen, for instance, antigen immobilized on a resin or surface, the bacteriophage will bind to the antigen. Bacteriophage which do not express the antibody will not bind to the antigen. Such panning techniques are well known in the art and are described for example, in Wright et al, (supra).
Processes, such as those described above, have been developed for the production of human antibodies using M13 bacteriophage display (Burton et al, 1994, Adv. Immunol. 57: 191-280). Essentially, a cDNA library is generated from mRNA obtained from a population of antibody -producing cells. The mRNA encodes rearranged immunoglobulin genes and thus, the cDNA encodes the same. Amplified cDNA is cloned into M13 expression vectors creating a library of phage which express human Fab fragments on their surface. Phage which display the antibody of interest are selected by antigen binding and are propagated in bacteria to produce soluble human Fab
immunoglobulin. Thus, in contrast to conventional monoclonal antibody synthesis, this procedure immortalizes DNA encoding human immunoglobulin rather than cells which express human immunoglobulin.
The procedures just presented describe the generation of phage which encode the Fab portion of an antibody molecule. However, phage which encode single chain antibodies (scFv/phage antibody libraries) are also useful in preparing Fab molecules useful in the invention. Fab molecules comprise the entire Ig light chain, that is, they comprise both the variable and constant region of the light chain, but include only the variable region and first constant region domain (CHI) of the heavy chain. Single chain antibody molecules comprise a single chain of protein comprising the Ig Fv fragment. An Ig Fv fragment includes only the variable regions of the heavy and light chains of the antibody, having no constant region contained therein. Phage libraries comprising scFv DNA may be generated following the procedures described in Marks et al, 1991, J. Mol. Biol. 222:581-597. Panning of phage so generated for the isolation of a desired antibody is conducted in a manner similar to that described for phage libraries comprising Fab DNA. Synthetic phage display libraries in which the heavy and light chain variable regions may be synthesized such that they include nearly all possible specificities (Barbas, 1995, Nature Medicine 1 :837-839; de Kruif et al, 1995, J. Mol. Biol. 248:97-105) may also be used to prepare an antibody useful in the practice of the invention.
Other methods for assessing the level of a protein include chromatography (e.g., HPLC, gas chromatography, liquid chromatography) and mass spectrometry (e.g., MS, MS-MS). For instance, a chromatography medium comprising a cognate receptor for the biomarker or a small molecule that binds to the biomarker can be used to substantially isolate the biomarker from the biological sample. Small molecules that bind specifically to a biomarker can be identified using conventional methods in the art, for instance, screening of compounds using combinatorial library methods known in the art, including biological libraries, spatially-addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the "one-bead one- compound" library method, and synthetic library methods using affinity chromatography selection.
The level of substantially isolated protein can be quantitated directly or indirectly using a conventional technique in the art such as spectrometry, Bradford protein assay, Lowry protein assay, biuret protein assay, or bicinchoninic acid protein assay, as well as immunodetection methods.
In another embodiment, the level of enzymatic activity of the biomarker if such biomarker has an enzymatic activity maybe quantified. Generally, enzyme activity may be measured by means known in the art, such as measurement of product formation, substrate degradation, or substrate concentration, at a selected point(s) or time(s) in the enzymatic reaction. There are numerous known methods and kits for measuring enzyme activity. For example, see US Patent No. 5,654, 152. Some methods may require purification of the Alzheimer's disease biomarker prior to measuring the enzymatic activity of the biomarker. A pure biomarker constitutes at least about 90%, preferably, 95% and even more preferably, at least about 99% by weight of the total protein in a given sample. Alzheimer's disease biomarkers of the invention may be purified according to methods known in the art, including, but not limited to, ion-exchange chromatography, size-exclusion chromatography, affinity chromatography, differential solubility, differential centrifugation, and HPLC. Determination of the Status of Alzheimer's Disease
The present invention is based on biomarker profiles or signatures determined for biological samples from individuals diagnosed with Alzheimer's Disease as well as from one or more other groups of control individuals (e.g., healthy control subjects not diagnosed with Alzheimer's Disease). The profile for Alzheimer's Disease was compared to the profile for biological samples from the one or more other groups of control individual. Those molecules differentially present, including those molecules differentially present at a level that is statistically significant, in the profile of Alzheimer's Disease samples as compared to another group (e.g., healthy control subjects not diagnosed with Alzheimer's Disease) were identified as biomarkers to distinguish those groups. The biomarkers are discussed in more detail elsewhere herein.
As described herein, assessment of results can depend on the statistical models used to predict the data. For example, AD biomarker levels relative to another reference level, which may be relative to the level of another AD biomarker, may be obtained according to an appropriate statistical model disclosed herein. In other methods described herein, high correlation amongst many biomarkers was observed to provide an indicator of disease state when analyzed using an appropriate statistical model.
The reference level used for comparison with the measured level for a AD biomarker may vary, depending on aspect of the invention being practiced, as will be understood from the foregoing discussion. For detection of AD, the "reference level" is typically a predetermined reference level, such as an average of levels obtained from a population that is not afflicted with AD, but in some instances, the reference level can be a mean or median level from a group of individuals including AD patients. In some instances, the predetermined reference level is derived from (e.g., is the mean or median of) levels obtained from an age-matched population. In some instances, the age-matched population comprises individuals with non-AD neurodegenerative disorders. In some instances, the reference level may be a historical reference level for the particular patient (e.g., a biomarker level that was obtained from a sample derived from the same individual, but at an earlier point in time). In some instances, the predetermined reference level is derived from (e.g., is the mean or median of) levels obtained from an age-matched population.
Age-matched populations (from which reference values may be obtained) are ideally the same age as the individual being tested, but approximately age-matched populations are also acceptable. Approximately age-matched populations may be within 1, 2, 3, 4, or 5 years of the age of the individual tested, or may be groups of different ages which encompass the age of the individual being tested. Approximately age-matched populations may be in 2, 3, 4, 5, 6, 7, 8, 9, or year increments (e.g. a "5 year increment" group which serves as the source for reference values for a 62 year old individual might include 58-62 year old individuals, 59-63 year old individuals, 60-64 year old individuals, 61-65 year old individuals, or 62-66 year old individuals.
The level(s) of the one or more biomarkers may be compared to
Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels using various techniques, including a simple comparison (e.g., a manual comparison) of the level(s) of the one or more biomarkers in the biological sample to Alzheimer's Disease- positive and/or Alzheimer's Disease-negative reference levels. The level(s) of the one or more biomarkers in the biological sample may also be compared to Alzheimer's Disease- positive and/or Alzheimer's Disease-negative reference levels using one or more statistical analyses.
Statistical models useful in the present invention includes but are not limited to Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Na'ive Bayes, Partial Least Squares (PLS), Random Forests, Shrunken Centroids, Sparse Partial Least Squares and Support Vector Machines approaches. Preferably, the statistical model is Partial Least Squares.
In some instances, the AD biomarkers of the invention include one or more of the biomarkers shown in Table 2. In one embodiment, the combination of biomarkers associated with assessing Alzheimer's disease is collectively presented on a detectable medium referred to as a panel or plasma based panel. Each of the biomarkers identified above may be used in concert with other biomarkers. For instance, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 10 or more, 15 or more, 20 or more, 30 or more, 40 or more, (and any integer value in between) Alzheimer's disease biomarkers may be used in concert with other, and with other known or to be known biomarkers for Alzheimer's disease.
In some instances, age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in identifying an individual for Alzheimer's disease. Therefore, in some instances, predictors for AD in an individual are selected from the group consisting of Alpha- 1 Microglobulin,
Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), and any combination thereof.
In other instances, predictors of AD in an individual are selected from the group consisting of Resistin, e3/e3, Pancreatic Polypeptide, e3/e4, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, 1-309, and any combination thereof.
In other instances, predictors of AD in an individual are selected from the group consisting of Cortisol, Pancreatic Polypeptide, osteopontin, IGF BP2, Resistin, and any combination thereof.
Based on the disclosure presented herein, a skilled artisan would understand that a plasma based signature for Alzheimer's disease can be detected in plasma and that signature can differentiate AD from healthy controls and other forms of dementia. The signature for Alzheimer's disease includes the combination of biomarkers disclosed herein. In some instances, the signature for Alzheimer's disease is a combination of biomarkers and predictors of Alzheimer's disease disclosed herein. For example, the biomarkers of the invention in combination with other factors such as age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), can improve diagnostic and screening accuracy. The biomarkers can also be combined with cognitive tests such as a simple memory test to improve diagnostic and screening accuracy. In some instances, the biomarkers of the invention can be combined with additional confirmatory CSF and imaging testing.
Additionally, Tables 2 provide a listing of biomarkers that are useful for identifying an individual with AD. In some instances, certain biomarkers are increases and other biomarkers are decreased in AD compared to age-matched normal controls (Tables 10, 11, and 12. Generally, a significant increase in a biomarker as compared to an appropriate control is indicative of AD, and a significant decrease in a biomarker as compared to an appropriate control is indicative of AD. In some instances, correlation between biomarkers provide an indication of AD in an individual. In some examples, any one or more of the biomarkers listed in Table 2 can be used to identify AD in an individual as distinguished from other non-AD individuals. The biomarkers of the invention can be used in diagnostic tests to assess the status of Alzheimer's disease in an individual, e.g., to diagnose Alzheimer's disease or to assess the degree of Alzheimer's disease in the individual. The phrase "Alzheimer's disease status" includes any distinguishable manifestation of the disease, including non- Alzheimer's disease, e.g., normal or non-demented. For example, disease status includes, without limitation, the presence or absence of Alzheimer's disease (e.g., Alzheimer's disease v. non-Alzheimer's disease), the risk of developing disease, the stage of the disease, the progress of disease (e.g., progress of disease or remission of disease over time) and the effectiveness or response to treatment of disease. Based on this status, further procedures may be indicated, including additional diagnostic tests or therapeutic procedures or regimens.
The ability of a diagnostic test to correctly predict status is commonly measured based on the sensitivity of the assay, the specificity of the assay or the area under a receiver operated characteristic ("ROC") curve. Sensitivity is the percentage of true positives that are predicted by a test to be positive, while specificity is the percentage of true negatives that are predicted by a test to be negative. An ROC curve provides the sensitivity of a test. The greater the area under the ROC curve, the more powerful the predictive value of the test. Other useful measures of the utility of a test are positive predictive value and negative predictive value. Positive predictive value is the percentage of people who test positive that is actually positive. Negative predictive value is the percentage of people who test negative that is actually negative.
As apparent from the example disclosed herein, diagnostic tests that use the biomarkers of the invention exhibit a sensitivity and specificity of at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% and about 100%. In some instances, screening tools of the present invention exhibits a high sensitivity of at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% and about 100%. Without wishing to be bound by any particular theory, it is believed that screening tools should exhibit high sensitivity, but specificity can be low. However, Diagnostics should have high sensitivity and specificity.
While individual biomarkers are useful diagnostic biomarkers, it has been found that a combination of biomarkers can provide greater predictive value of a particular status than single biomarkers alone. Specifically, the detection of a plurality of biomarkers in a sample can increase the sensitivity and/or specificity of the test. A combination of at least two biomarkers is sometimes referred to as a "biomarker profile" or "biomarker fingerprint." Examples of Alzheimer's disease biomarkers of the invention include, but are not limited, to Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C,
Thrombomodulin, TIMP-1, VCAM-1, VEGF, Von Willebrand Factor, and any combination thereof. In some instances, age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) are additional factors that are considered in evaluating the diagnosis of an individual for Alzheimer's disease.
In addition, the methods disclosed herein using the biomarkers listed in the tables presented herein may be used in combination with clinical diagnostic measures of Alzheimer's Disease and/or other neurodegenerative diseases. Combinations with clinical diagnostics may facilitate the disclosed methods, or confirm results of the disclosed methods (for example, facilitating or confirming diagnosis, monitoring progression or regression, and/or determining predisposition to Alzheimer's Disease).
Determining Alzheimer's disease status typically involves classifying an individual into one of two or more groups based on the results of the diagnostic test. The diagnostic tests described herein can be used to classify an individual into a number of different states. In one embodiment, the invention provides methods for determining the presence or absence of Alzheimer's disease in an individual (status: Alzheimer's disease v. non-Alzheimer's disease). The presence or absence of Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers in samples obtained from individuals and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular risk level.
In another embodiment, the invention provides methods for determining the risk of developing disease in an individual. Biomarker amounts or patterns are characteristic of various risk states, e.g., high, medium or low. The risk of developing Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers in sample obtained from individuals and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular risk level.
In yet another embodiment, the invention provides methods for determining the stage of Alzheimer's disease in an individual. Each stage of the disease can be characterized by the amount of a biomarker or relative amounts of a set of biomarkers (i.e., a pattern) that are found in a sample obtained from the individual. The stage of Alzheimer's disease is determined by measuring the relevant biomarker or biomarkers and then either submitting them to a classification algorithm or comparing them with a reference amount and/or pattern of biomarkers that is associated with the particular stage.
In another embodiment, the invention provides methods for determining the course of Alzheimer's disease in an individual. Disease course refers to changes in disease status over time, including disease progression (worsening) and disease regression (improvement). Over time, the amounts or relative amounts (e.g., the pattern) of the biomarkers changes. For example, levels of various biomarkers of the present invention increase with progression of disease. Accordingly, this method involves measuring the level of one or more biomarkers in an individual at two or more different time points, e.g., a first time and a second time, and comparing the change in amounts. The course of disease is determined based on these comparisons.
In some instances, the levels of various biomarkers of the invention decreases with disease progression. In this method, the level of one or more biomarkers in a sample from an individual is measured at two or more different time points, e.g., a first time and a second time, and the change in levels, if any is assessed. The course of disease is determined based on these comparisons.
Similarly, changes in the rate of disease progression (or regression) may be monitored by measuring the level of one or more biomarkers at different times and calculating the rate of change in biomarker levels. The ability to measure disease state or rate of disease progression is important for drug treatment studies where the goal is to slow down or arrest disease progression using therapy.
Additional embodiments of the invention relate to the communication of the results or diagnoses or both to technicians, physicians or patients, for example. In certain embodiments, computers are used to communicate results or diagnoses or both to interested parties, e.g., physicians and their patients.
In certain embodiments, the methods of the invention further comprise managing individual treatment based on their disease status. Such management includes the actions of the physician or clinician subsequent to determining Alzheimer's disease status. For example, if a physician makes a diagnosis of Alzheimer's disease, then a certain regime of treatment, such as prescription or administration of the therapeutic compound might follow. Alternatively, a diagnosis of non-Alzheimer's disease might be followed by further testing to determine any other diseases that might the patient might be suffering from. Also, if the test is inconclusive with respect to Alzheimer's disease status, further tests may be called for.
In a preferred embodiment of the invention, a diagnosis based on the presence or absence or relative levels in the biological sample of an individual of the relevant biomarkers disclosed herein is communicated to the individual as soon as possible after the diagnosis is obtained.
According to yet another aspect, the present invention provides a method of assessing efficacy of a treatment of Alzheimer's disease in a patient comprising: a) determining a baseline level of biomarkers in a first sample obtained from the patient before receiving the treatment; b) determining the level of same biomarkers in a second sample obtained from the patient after receiving the treatment; wherein an alteration in the levels of the biomarkers in the post-treatment sample is correlated with a positive treatment outcome.
Assays
The experiments disclosed herein were designed to develop an assay to identify universal accepted biomarkers for diagnosing, screening, monitoring and staging neurodegenerative diseases such as Alzheimer's disease that are fast, more accurate, and less expensive. The results presented herein demonstrate that a diagnostic assay has been developed that can detect among other things early onset of Alzheimer's disease.
Detection of early onset of Alzheimer's disease is believed to increase the success rate of the individual being successfully treated for Alzheimer's disease.
The diagnostic method of the present invention can be applied to subjects who have been previously diagnosed with Alzheimer's disease, those who are suspected of having Alzheimer's disease, and those at risk of developing Alzheimer's disease. For example, patients diagnosed with dementia, in particular, those patients who were previously clinically normal, are suitable subjects. However, it is not intended that the present invention be limited to use with any particular subject types.
According to some embodiments, the subject is a human subject.
According to certain embodiments, the subject is selected from the group consisting of subjects displaying pathology resulting from Alzheimer's disease, subjects suspected of displaying pathology resulting from Alzheimer's disease, and subjects at risk of displaying pathology resulting from Alzheimer's disease.
According to another embodiment, the Alzheimer's disease diagnosed using the method of the present invention is selected from the group consisting of late onset Alzheimer's disease, early onset Alzheimer's disease, familial Alzheimer's disease and sporadic Alzheimer's disease.
Early-onset Alzheimer's disease (EOAD) is a rare form of Alzheimer's disease in which individuals are diagnosed with the disease before age 65. Less than 10% of all Alzheimer's disease patients have EOAD. Younger individuals who develop Alzheimer's disease exhibit more of the brain abnormalities that are normally associated with Alzheimer's disease. EOAD is usually familial and follows an autosomal dominant inheritance pattern. To date, mutations in three genes including amyloid precursor protein (APP) on chromosome 21, presenilin 1 (PSEN1) on chromosome 14 and presenilin 2 (PSEN2) on chromosome 1 have been identified in families with EOAD . Mutations in the APP, PSEN1 and PSEN2 genes account for about 50% of the disease . Most of the pathogenic mutations in the APP and presenilin genes are associated with abnormal processing of APP, which leads to the overproduction of toxic Αβ-1-42. Down syndrome patients, who have three copies of chromosome 21 which includes the APP gene, begin to develop the characteristic senile plaques and tau tangles at the ages of 30 and 40 (M. Ilyas Kamboh (2004), Molecular Genetics of Late-Onset Alzheimer's Disease, Annals of Human Genetics 68(4):381-404).
Late-onset Alzheimer's disease (LOAD) is the most common form of Alzheimer's disease, accounting for about 90% of cases and usually occurring after age 65. LOAD strikes almost half of all individuals over the age of 85 and may or may not be hereditary. It is a complex and multifactorial disease with the possible involvement of several genes. Genome-wide linkage or linkage disequilibrium studies on LOAD have provided informative data for the existence of multiple putative genes for Alzheimer's disease on several chromosomes, with the strongest evidence on chromosomes 12, 10, 9 and 6. LOAD cases tend to be sporadic, wherein there is no family history of the disease. Genetic susceptibility at multiple genes and interaction between these genes as well as environmental factors are most likely responsible for the etiology of LOAD. Twin data on incident cases indicates that almost 80% of the LOAD risk is attributable to genetic factors. The Apolipoprotein E (APOE) gene on chromosome 19ql3 has been identified as a strong risk factor for LOAD. In fact, the ΑΡΟΕ-ε4 allele has been established as a strong susceptibility marker that accounts for nearly 30% of the risk in late-onset Alzheimer's disease. More specifically, three variants of APOE, encoded by codons 1 12 and 158, have been found to modify the risk of LOAD. As compared to the common APOE-83 allele (codon 1 12=Cys and codon 158=Arg), the ΑΡΟΕ-ε4 allele (codon 112=Arg and codon 158=Arg) increases the risk of Alzheimer's disease, while the ΑΡΟΕ-ε2 allele (codon 1 12=Cys and codon 158=Cys) decreases the risk of Alzheimer's disease. The effect of the ΑΡΟΕ-ε4 allele is dose related, wherein one or two copies of the ΑΡΟΕ-ε4 allele are associated with 3-fold or 15-fold risk, respectively. However, the effect of the ΑΡΟΕ-ε4 allele on Alzheimer's disease risk appears to decline with increasing age (M. Ilyas Kamboh (2004), supra).
Based on the disclosure presented herein, a skilled artisan would understand that a profile of biomarkers for Alzheimer's disease can be detected in plasma and that profile can differentiate AD from healthy controls and other forms of dementia. The profiles for Alzheimer's disease includes the biomarkers disclosed herein. In some instances, the profile for Alzheimer's disease is a combination of biomarkers and other factors of Alzheimer's disease disclosed herein. For example, the biomarkers of the invention in combination with other factors such as age, gender, ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4), can improve diagnostic and screening accuracy. The biomarkers can also be combined with cognitive tests such as a simple memory test to improve diagnostic and screening accuracy. In some instances, the biomarkers of the invention can be combined with additional confirmatory CSF and imaging testing.
For example, the biomarkers of the invention can be combined with existing criteria for dementia to improve diagnostic and screening accuracy of
Alzheimer's disease. Dementia is the decline of memory and other cognitive functions in comparison with the patient's previous level of function as determined by a history of decline in performance and by abnormalities noted from clinical examination and neuropsychological tests. A diagnosis of dementia cannot be made when consciousness is impaired by delirium, drowsiness, stupor, or coma or when other clinical abnormalities prevent adequate evaluation of mental status. Dementia is a diagnosis based on behavior and cannot be determined by computerized tomography, electroencephalography, or other laboratory instructions, although specific causes of dementia may be identified by these means. In some instances, the biomarkers of the invention can be combined with existing criteria Alzheimer's disease. A clinical diagnosis of probable Alzheimer's disease can be made with confidence if there is a typical insidious onset of dementia with progression and if there are no other systemic or brain diseases that could account for the progressive memory and other cognitive deficits. Among the disorders that must be excluded are manic depressive disorder, Parkinson's disease, multiinfarct dementia, and drug intoxication; less commonly encountered disorders that may cause dementia include thyroid disease, pernicious anemia, luetic brain disease and other chronic infections of the nervous system, subdural hematoma, occult hydrocephalus, Huntington's disease, Creutzfeldt- Jakob disease, and brain tumors.
A diagnosis of definite Alzheimer's disease requires histopathologic confirmation. A clinical diagnosis of possible Alzheimer's disease may be made in the presence of other significant diseases, particularly if, on clinical judgment, Alzheimer's disease is considered the more likely cause of the progressive dementia. The clinical diagnosis of possible rather than probable Alzheimer's disease may be used if the presentation or course is somewhat aberrant. The information needed to apply these criteria is obtained by standard methods of examination: the medical history; neurologic; psychiatric, and clinical examinations; neuropsychological tests; and laboratory studies.
It is believed that some of the biomarkers for Alzheimer's Disease described herein may also be biomarkers for neurodegenerative diseases in general.
Therefore, it is believed that at least some of the Alzheimer's Disease biomarkers may be used in the methods described herein for neurodegenerative diseases in general. That is, the methods described herein with respect to Alzheimer's Disease may also be used for diagnosing (or aiding in the diagnosis of) a neurodegenerative disease, methods of monitoring progression/regression of a neurodegenerative disease, methods of assessing efficacy of compositions for treating a neurodegenerative disease, methods of screening a composition for activity in modulating biomarkers associated with a neurodegenerative disease, methods of identifying potential drug targets for neurodegenerative diseases, and methods of treating a neurodegenerative disease. Such methods could be conducted as described herein with respect to Alzheimer's Disease.
Kits
A kit is envisaged for every method disclosed. The following description of a kit useful for diagnosing Alzheimer's disease in an individual by measuring the level of a biomarker in a biological sample therefore is not intended to be limiting and should not be construed that way.
The kit may comprise a negative control containing a biomarker at a concentration of about the concentration of the biomarker which is present in a biological sample of an individual who does not have Alzheimer's disease or does not have increased risk for Alzheimer's disease. The kit may also include a positive control containing the biomarker at a concentration of about the concentration of the biomarker which is present in a biological sample of an individual who as Alzheimer's disease or has increased risk for Alzheimer's disease.
Additionally, the kit includes a panel of biomarkers including one or more of the biomarkers shown in Table 2. However, the invention should not be limited to only these markers disclosed herein (e.g., Table 2) because a skilled artisan when armed with the present disclosure would be able identify additional markers that can be used as indicators for Alzheimer's disease. For example, as discussed in the Examples, a test sample and a control sample can be subjected to any commercially available panel comprising a plurality of markers and analyzed according to the statistic models disclosed herein to identify markers associated with AD.
Moreover, the disclosure presented here demonstrates a high degree of correlation amongst certain markers for identifying AD in an individual. For example, NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an AD specific pathophysiological pathway. Without wishing to be bound by any particular theory, it is believed that analytes that exhibit high correlation can be interchanged with other highly correlated analytes and still obtain similar differentiation performance. Thus, markers that are associated with an AD specific pathophysiological pathway can be interchangeable. Accordingly, correlation amongst the markers of the invention provides means to identify other related markers associated with the specific pathophysiological pathway as being indicators for
Alzheimer's disease.
One or more of the biomarkers in each or all of Tables 2, 3, 7, 8, 9, 10, 11, 12 or any fraction thereof can be included in the kit. In another aspect, other factors that predict for of AD can be included in the kit. Such factors include but are not limited to ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4).
In one embodiment, the kit comprises a panel comprising at least Cortisol, Pancreatic Polypeptide, osteopontin, IGF BP2, and Resistin. In another embodiment, the kit comprises a panel comprising at least Resistin, e3/e3, Pancreatic Polypeptide, e3/e4, ApoD, G-CSF, MlP lbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, e2/e3, MMP 1, 1-309.
In another embodiment, the kit comprises a panel comprising Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, BLC, Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP, Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, TIMP-1, VCAM-1, VEGF, and Von Willebrand Factor.
In another embodiment, the kit comprises reagents to assess age, gender,
ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual.
The kit of the invention can be used to assess the status of Alzheimer's disease in an individual, e.g., to diagnose Alzheimer's disease or to assess the degree of Alzheimer's disease in the individual. The phrase "Alzheimer's disease status" includes any distinguishable manifestation of the disease, including non-Alzheimer's disease, e.g., normal or non-demented. For example, disease status includes, without limitation, the presence or absence of Alzheimer's disease (e.g., Alzheimer's disease v. non-Alzheimer's disease), the risk of developing disease, the stage of the disease, the progress of disease (e.g., progress of disease or remission of disease over time) and the effectiveness or response to treatment of disease. Based on this status, further procedures may be indicated, including additional diagnostic tests or therapeutic procedures or regimens.
Furthermore, the kit includes an instructional material for use in the diagnosis of Alzheimer's disease in an individual. The instructional material can be a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the method of the invention in the kit for assessment of Alzheimer's disease risk in a individual. The instructional material of the kit of the invention may, for example, be affixed to a container which contains other contents of the kit, or be shipped together with a container which contains the kit. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the contents of the kit be used cooperatively by the recipient.
EXAMPLES The invention is further described in detail by reference to the following experimental examples. These examples are provided for purposes of illustration only, and are not intended to be limiting unless otherwise specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.
The experiments disclosed herein were designed to develop an assay to identify universal accepted biomarkers for diagnosing, monitoring and staging neurodegenerative diseases such as Alzheimer's disease that are fast, more accurate, and less expensive. The results presented herein demonstrate that a diagnostic assay has been developed that can detect among other things early onset of Alzheimer's disease.
Example 1 : Assessing diagnostic accuracy of a plasma based multiplexed immunoassay panel to identify Alzheimer's disease
Currently, prodromal Alzheimer's can be identified through the use of cognitive tests combined with cerebrospinal fluid (CSF) and imaging biomarkers.
However, such tests are often costly and invasive and are difficult to implement as screening tools to identify individual s suitable for clinical trial study. The identification of a screening blood test would significantly enable prevention studies in early
Alzheimer's disease. The present study examines the suitability of a multiplexed plasma panel as a screening tool to differentiate Alzheimer's disease from age-matched controls.
Briefly, a luminex based multiplex immunoassay panel consisting of 190 analytes was utilized in plasma samples derived from the University of Pennsylvania. The strategy to identify a plasma based panel included multivariate based approaches combined with examining changes in both CSF in both autopsy confirmed and non- autopsy confirmed individuals from the UPenn biobank. Final analytes were chosen based upon high performance in multivariate models and upon expression in CSF from autopsy confirmed individuals.
Multivariate training/test set models using all 190 analytes suggested Alzheimer's disease individuals could be differentiated from controls using analytes on the panel. Partial least square and shrunked centroid algorithms exhibited the best performance. Many of the top analytes identified in the multivariate and ANOVA analysis were also expressed in CSF obtained from autopsy confirmed individuals.
Multivariate and CSF results were combined to generate a final panel consisting of 24 multiplex analytes plus age, gender and ApoE genetic status (6 co-variates e2/e2; e2/e3; e2/e4; e3/e4; e3/e3; e4/e4). Performance of the panel in differentiating Alzheimer's disease from controls in the entire UPenn sample set using a logistic regression analysis exhibited 80% sensitivity 80% specificity.
Multivariate approaches combined with CSF-plasma expression and pathway analysis were utilized to identify a 32 variate algorithm that differentiates Alzheimer's disease from age-matched controls with high sensitivity suitable for preliminary screening of individuals who might be eligible for clinical trial enrollment in early Alzheimer's disease studies.
The materials and methods employed in these experiments are now described.
University of Pennsylvania Sample Set
Individuals were recruited and longitudinally followed at the University of Pennsylvania (UPenn). All protocols were approved by the University of Pennsylvania Review Board and all individuals, and in cases of dementia individuals, caregivers, provided an informed consent for utilization of biofluids for exploratory research purposes. Clinical information was de-identified in compliance with privacy regulations. Individuals diagnosed with clinical Alzheimer's disease and frontotemporal lobar degeneration (FTLD), progressive supranuclear palsy (PSP) and corticobasal syndrome (CBS) were longitudinally followed in the ALS center and Frontotemporal dementia clinic. Each individual underwent detailed neurological and laboratory examination to ensure the accuracy of clinical diagnosis according to established criteria for Alzheimer's disease and FTLD. Plasma samples were obtained from 92 Alzheimer's disease individuals, 126 normal healthy controls and 48 with other forms of dementia. Autopsy confirmed CSF samples were obtained from 70 Alzheimer's disease individuals and 25 individuals with other dementia (TDP, Tau and DLB). 32 CSF samples from cognitively normal, non-autopsied elderly controls were also tested. ApoE genotyping
ApoE genotyping was done for all individuals using EDTA blood samples. Taqman quantitative PCR assays were used for genotyping APOE nucleotides 334 T/C and 472 C/T with an ABI 7900 real time thermocylcer using DNA freshly prepared from EDTA whole blood. Luminex xMAP panel
Plasma samples were analyzed using a commercially available multiplexed luminex human discovery xMAP panel from Rules Based Medicine (RBM, Austin, Texas). All assays were validated according to CLIA standards. In brief, the luminex technology multiplexes immunoassays on the surface of polystyrene microsphere beads. The microsphere beads are loaded with a ratio of two spectrally distinct flourochromes yielding up to 100 uniquely color-coded beads. The beads are coated with capture antibodies specific for the assay and run in either standard sandwich or competitive immunoassay format. Capture-antibody microspheres are incubated with blocking solution and diluted plasma sample or calibration controls for one hour. Beads are rinsed and biotinylated detection reagent added. Streptavidin-phycoerthyrin is then added to each well and incubated for 60 minutes. Following wash steps, beads are resuspended in reading solution and read on the luminex instrument. Details of each assay procedure are provided by RBM in validation reports that are compliant with CLIA standards. Some of the assays have defined a lower limit of quantitation. For the purposes of the present study, the lower limit of detection was utilized. Validation details of all the assays are available from Rules Based Medicine (http://www.rulesbasedmediciiie.com). Plasma Collection Methods
In brief, whole blood was collected into 10ml BD lavender top K2EDTA(18mg spray coated or 15% solution) coated vacutainers. Samples were gently mixed by inversion and spun at 3000 rmp for 15 minutes at room temperature within one hour of collection. Immediately after centrifugation, plasma was transferred to a polypropylene transfer tube, placed in dry ice and allowed to completely freeze. Samples were then stored at -80C° until analysis. A 0.5ml aliquot that had not been allowed any freeze-thaw cycles was shipped to RBM for analysis.
Statistical Methods
For purposes of the univariate analysis, analytes that had more than 10% missing values were excluded from the analysis. Values that were reported as LOW (below the lowest assay limit which is defined by RBM as a value below the lowest calibrator for the individual assay) were imputed by taking the reported low detection limit for individual assays (LDD) and dividing by 2. Individual analyte distributions were tested for normalcy using Box-Cox analysis. If necessary, analytes were log transformed and checked again for normalcy. The majority of analytes exhibited non-normal distributions and were log transformed. For purposes of the univariate analysis, multidimensional scaling and Mahalanobis distances were used to detect outliers. Outliers were defined as 5 standard deviations beyond the mean and were replaced with the 5 STD value. A major concern in the analysis was control of type I error rate due to relatively large number of plasma proteins in the multiplex panel. False discovery (FDR) corrections were applied to p-values to account for the multiple comparisons. ANOVA models including diagnosis, age, gender and ApoE4 allele status were utilized in the final model. ApoE allele status was classified into 6 subcategories; e2/e2, e2/e3, e2/e4, e3/e3, e3/e4, e4/e4.
Multiple marker analysis approach was used to build relationships between disease groups. Approaches applied included logistic regression, boosted tree models, FDA, KNN, Naive Bayes, Partial Least Squares, Random Forests, shrunken centroids, sparse partial least squares and support vector machine approaches. Three models were attempted using different co-variates: 1) age, gender and ApoE4 genetic status alone, 2) RBM assay results only and 3) RBM assay results plus age, gender and ApoE4 allele status in order to understand the predictive ability of the assays beyond demographic and genotype information currently used to assess risk. Prior to modeling, pre-filtering of markers using K resampling iterations in an unsupervised fashion were performed on UPenn dataset. This included splitting UPenn data into training and test sets and then applying an unsupervised filter on the predictors. The predictive model using only UPenn data was further built and tuned on the training set. The model was then tested on the UPenn test dataset as an independent dataset separate from the trained dataset.
Classification accuracy and kappa features of the performance were saved. All feature selection routines were extensively cross validated using methods described by Ambroise and McLachlan 2002 PNAS). Measures of marker importance were biased towards those that use uncertainty (logistic regression slope tests) as opposed to those that did not (random forest variable importance etc).
The results of the experiments are now described.
Demographics of the UPenn cohort
Table 1 summarizes demographic characteristics of the UPenn sample set. Two hundred and sixty six plasma samples were shipped for multiplex analysis. Of the 266, 92 were from Alzheimer's disease, 126 were from healthy elderly controls and 48 were from individuals with other forms of dementia. Control and dementia individuals from the UPenn cohort were significantly younger than Alzheimer's disease. Thus age was included as a co-variate in all models. Amongst the groups, females were more highly represented, but there were no significant differences in gender ratios between the groups. Gender was also included as a co-variate in all the models. The prevalence of ApoE4 allele in the Alzheimer's disease group was 63%, very similar to what was observed in general Alzheimer's disease population. In addition, 102 CSF samples from autopsy confirmed individuals and 32 CSF samples from non-autopsy confirmed CSF controls were included in the analysis to determine whether changes in plasma analytes were reflected by changes in CSF from autopsy confirmed individuals. Age, gender and ApoE prevalence did not differ between the plasma and CSF cohorts.
Table 1 : Demographics University of Pennsylvania Sample Set
Figure imgf000052_0001
*p<0.05 when compared to Alzheimer's disease
Table 2 summarizes all the analytes included in the RBM panel. Light gray highlights those analytes that had more than 10% missing or had more than 10% LOW values. Multivariate approaches were combined with CSF expression patterns for feature selection. The top 24 performers are highlighted in dark gray (Table 2). The multivariate analyses in this context are the predictive models that were performed (e.g. Random forest, PLC, etc.) for the goal of predicting the outcome.
Table 2: Analyte and final model summary. Analytes with more than 10% missing or had more than 10% LOW values (highlighted in light gray). Final panel (highlighted in dark grey) were selected based upon multivariate results and upon expression from auto sy confirmed individuals.
Figure imgf000053_0001
Figure imgf000054_0001
A number of multivariate approaches were applied to aid in final feature selection. The strategy for model generation relied upon training with 75% of data and then testing on 25% of the remaining cohort data. Multivariate strategies included a shrunken centroid algorithm developed for predictive analysis of microarrays (PAM) (Tusher et al, 2001 Proc Natl Acad Sci U S A. 98(9):5116-5121), FDA, KNN, logistic regression, Naive Bayes, partial least squares, random forest, sparse partial least square, boosted trees, linear discriminant analysis (data not shown), principal components (data not shown) and support vector machine (Vapnik et al, 1999 IEEE Trans Neural Netw. 10(5):988-999). ROC curves from the analysis are shown in Figure 1. Partial least squares and shrunken centroid approaches appeared to give maximal performance in the training and test sets. Because the cohorts were biased in terms of enrollment, all models included age, gender and ApoE4 allele status as co-variates. Figure 1 lists the top 30 contributors to the partial least square model. Genotype was an important contributor and appeared in the top list. As a result, genotype models were used as baseline models for subsequent comparisons.
ANOVA analysis were then conducted on all the analytes to determine whether changes were statistically significant between Alzheimer's disease and age- matched controls and Alzheimer's disease and other dementias. Although many analytes were statistically different in the Alzheimer's disease group compared to either control or other dementia groups (Table 3), there were few analytes that exhibited robust differences (e.g. more than 1.5X) compared to the Alzheimer's disease group. Interestingly, a subset of analytes appeared to differentiate Alzheimer's disease from other types of dementias including a number of the apolipoproteins and inflammatory factors.
Table 3 : Summary of analyte levels in UPenn Plasma and ANOVA Results: Age and gender included as co-variates and p values reported for Alzheimer's disease v control
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
0 8.52 + 2.60 0 7.23 1.51
Π- Ι 2ρ40 Alzheimer' s disease 0.0054 Thrombospon Alzheimer's disease 0.0491 <ng ml) 0.53 + 0.25 din- 1 (ng.ml) 293.2 + 11228.5 <0.0001*
C 0.44 + 0.23 C 7581.5 + 9044.7
0 0.46 + 0.23 0 14217.2 + 12022.1
II .- 1 (pg'ml) Alzheimer's disease 0.0448 VK(il Alzheimer's disease 0.0956
101.16 + 41.82 0.0013* (pg'ml) 605.57 + 316.20
C 88.69 + 35.28 C 531.61 + 267.38
0 78.98 + 34.87 0 607.14 + 248.07
II - I ra (pg'ml) Alzheimer's disease 0.7702 Von Alzheimer's disease 0.3465
85.64 + 66.19 0.0010* Willelirand 33.04 + 18.23 0.0193* C 82.15 + 51.83 1 -'actor C 28.13 + 13.68 0 121.88 + 90.69 (ug ml) 0 36.24 + 16.29
A comparison analysis was then conducted to examine expression in CSF from autopsy confirmed individuals with analytes that exhibited statistical significance by ANOVA analysis or were identified as top differentiators in the multivariate models. Figure 2 summarizes expression of a subset.
A logistic regression model was utilized to compare performance of a baseline model (age, gender and ApoE allele status) to a model including only the 24 top plasma analytes and a model including all 24 plasma analytes plus the baseline model. All models were benchmarked at 80% sensitivity to allow comparisons across the models. In general, the baseline demographic model using only age, gender and ApoE4 allele showed positive predictive value of 52%. A model including all 24 analytes showed a positive predictive value of 64% and a model including all 24 analytes and baseline demographics showed a reasonable positive predictive value of 75% (see Table 4). ROC curves for each of the models are depicted in Figure 3. The final logistic regression model including age, gender, ApoE allele and 24 plasma analytes yielded a 80% sensitivity and 80% specificity in differentiating Alzheimer's disease from controls. Model showed very good performance in differentiating Alzheimer's disease from other forms of dementia as well. A model was attempted using 12 of the 18 analytes described by Ray et al. (2007 Nat Med. 13(1 1): 1359-1362) (Angiopoietin-2, EGF, G-CSF, ICAM- 1, Interleukin 3, Interleukin 8/CXCL8, M-CSF, PDGF-BB, PARC/CCL18,
RANTES/CCL5 and TNF-alpha). Two analytes (Interleukin la, Interleukin- 11) were below the limit of detection and not included in the analysis. Four analytes
(MIP5/CCL15, GDNF, IGF-BP6, TRAIL-R4), were not present on the panel, but related analytes including IGF-BP2 and TRAIL R3 were represented. Internal studies using the Ray et al. (2007 Nat Med. 13(1 1): 1359-1362), data and excluding MIP5, GDNF, IGF- BP6 and TRAIL-R4 did not degrade the performance of the model (data not shown). Preliminary results could not confirm performance reported in Ray et al. (2007 Nat Med. 13(1 1): 1359-1362), using multiplex analytes alone (data not shown). However, inclusion of demographic information improved performance close to levels of the 32 co-variate model described in Figure 3.
Figure imgf000059_0001
Figure imgf000059_0002
Est mates enc mar e aga nst sens t v ty n a mo e s
Correlation matrix analysis in the Alzheimer's disease samples showed a high degree of correlation amongst the 24 analytes (Figure 4A). Figure 4B is a chart also demonstrating high correlation of the analytes with each other and that there is high correlation with other analytes on the panel. Without wishing to be bound by any particular theory, it is believed that any of the individual 24 analyte can be switched out with any analyte that showed significant correlation with another analyte on the panel (p<0.05). For example, NT-proBNP showed high correlation with IGFBP2 and beta2 microglobulin suggesting these analytes may be linked in an Alzheimer's disease specific pathophysiological pathway. Many of the analytes in the 190 panel showed high correlation and it was possible to interchange the top 24 analytes with other highly correlated analytes and obtain similar differentiation performance (data not shown).
A subanalysis was completed to examine the contribution of ApoE genotype to multiplex analyte expression. There has been some controversy in the literature around whether individuals with an ApoE4 allele exhibit altered protein levels. In the current dataset, ApoE plasma protein levels differed based upon ApoE allele genotype irrespective of diagnostic status. For example, ApoE4/E4 homozygotes showed low levels of plasma ApoE protein compared to ApoE2/E2 homozygotes (Figure 5). The phenomena was not limited to ApoE protein levels as a similar pattern was noted in plasma C Reactive Protein levels (Figure 3). IL-15 levels were elevated in ApoE4/E4 homozygotes. These data suggest an ApoE endophenotype is associated with specific patterns of expression of apolipoprotein and cytokine analytes irrespective of diagnosis.
Figure 5 compares apolipoprotein E levels in individuals as a function of their ApoE genotype. In both datasets, individuals with either an e3/e4 or e4/e4 genotype showed lower apolipoprotein levels compared to individuals with e2/e2, e3/e2 or e3/e3 genotypes. Individuals with an e2/e4 genotype exhibited levels more closely matched to e3 and e2 groups. Although the numbers are very small, individuals homozygous for E2 exhibited much higher protein levels. Alzheimer's disease individuals were present in each of the ApoE genetic groups with the exception of e2 homozygotes. These data suggest that apolipoprotein levels do differ dependent upon ApoE genotype and that levels are lowest in individuals with an e3/e4 or and e4/e4 genotype.
Plasma Biomarkers for Diagnosis of Alzheimer's disease
Identification of Alzheimer's disease individuals prior to the onset of dementia will be critical for successful development of Alzheimer's disease disease modifying drug. Although there are CSF and imaging research tools currently available to identify individuals at risk of progressing to dementia, these tools are difficult to implement as screening tools for individual enrollment due to the cost and invasive nature of CSF testing. A simple blood test would aid in identifying those eligible for more confirmatory biomarker and cognitive testing and would help raise awareness around dementia prevention.
The current study examined the utility of a commercially available multiplex blood panel in differentiating Alzheimer's disease from controls using samples obtained from the University of Pennsylvania. Initial studies were initiated based upon a report by Ray et al, (2007 Nat Med. 13(11): 1359-1362) that an 18 analyte panel could differentiate Alzheimer's disease from controls. Performance results of the a subset of the Ray et al. (2007 Nat Med. 13(1 1): 1359-1362), analytes in the UPenn sample set could only be reproduced if age and ApoE genetic allele status were included in the model suggesting performance was largely driven by age and ApoE status.
Multiple approaches were used for feature selection from the 190 analyte.
The top lists of obtained from multivariate approaches were compared to ANOVA lists and lists obtained from examining expression of analytes in CSF from autopsy confirmed individuals. A final model containing 24 analytes was selected for further analysis. Inclusion of the 24 multiplex analytes improved performance over baseline demographic models in identifying individuals with Alzheimer's disease. Logistic regression estimates of model performance showed 80% sensitivity and 80% specificity in differentiating Alzheimer's disease from controls. However, caution should be exercised as these estimates are likely to degrade in a population setting and when tested in independent datasets. Correlation analysis utilized in the current study to identify analyte redundancy in the panel was actually quite revealing as many of the analytes showed strong correlations. The finding may be indicative of a biological signaling cascade activated during pathological sequeale associated with dementia. Further pathway analysis may shed insight into the underlying pathophysiological and specific endophenotypes associated with Alzheimer's disease and other forms dementia.
Finally, the current study demonstrated that plasma apolipoprotein E levels are altered dependent upon genotypic status. Specifically, individuals with an e3/e4 or e4/e4 genotype exhibited lower apolipoprotein E and C reactive protein and higher IL-14 levels compared to other ApoE genotypes. Reports of altered ApoE protein levels in Alzheimer's disease individuals have been controversial and studies examining changes in message levels often contradict protein findings (Kim et al, 2009 Neuron 63(3):287- 303). Apolipoprotein E is a glycoprotein involved in lipid transport and a ligand for receptor mediated endocytosis. It is one of the major high density lipid (HDL) components in brain and believed to be critical for cholesterol transport involved in brain synaptic turnover. ApoE is the only gene consistently shown to increase the risk of late onset Alzheimer's disease and E4 is believed to confer greater risk while E2 is believed to be protective for dementia. The finding that ApoE4 levels are lower in plasma in Alzheimer's disease individuals may have relevance to disease if levels are altered in a similar way in CNS. Low levels may impact normal synaptic function and membrane recycling at the synapse (Hirsch-Reinshagen et al., 2009 Mol Cell Biochem. 326(1- 2): 121-129). Interestingly, a novel genetic risk factor has been identified in another apolipoprotein, ApoJ also known as clusterin. The current study did not identify any changes in ApoJ levels in either cohort and additional studies in CSF are needed to further understand relevance.
The current findings support that a plasma based signature for Alzheimer's disease can be detected in plasma and that signature can differentiate Alzheimer's disease from healthy controls and other forms of dementia. The signature can improve diagnostic accuracy over age and ApoE4 allele status alone, but does not rise to the level of a standalone diagnostic tool. The panel may, when combined with a simple memory test, have utility as a screening tool for individuals with a cognitive complaint who would then be eligible for more confirmatory CSF and imaging testing.
Numerous proteomic approaches have long sought a diagnostic signature in plasma or serum (Ray et al, 2007 Nat Med. 13(1 1): 1359-1362, Hye et al, 2006 Brain 129(Pt l l):3042-3050; Irizarry et al, 2004 NeuroRx. l(2):226-234; Song et al, 2009 Brain Res Rev.; Zhang et al, Proteomics 4(l):244-256). Unfortunately, much of the current proteomic technological approaches have been qualitative in nature and more quantitative confirmation in independent studies using more confirmative immunoassay approaches have been elusive. Here, a quantitative approach was utilized to assess performance of a multiplex panel in differentiating Alzheimer's disease from controls. The panel showed a significant, improvement over use of age, gender and ApoE4 allele status alone.
Example 2: RBM Biomarker Analysis
The data set consisted of 165 characteristics (i.e. variables) collected on
218 individuals. Of these individuals, 92 were determined to be Alzheimer's disease and
126 were controls.
The following variables were removed due to missing values or other factors: IgE, IL12p70, IL17E, IL1 alpha, Prostate Specfic Ag, Tissue Factor. After filtering, there were 164 variables used for prediction. The data were split into a training set (n=175) and a test set(n=43) in a manner that preserves the frequency distribution between Alzheimer's disease and control individuals. Where needed, the training and test sets were centered and scaled. Models
A variety of statistical models were used to predict the data:
• Random Forests: A sequence of 3 values for the number of retained variables were used to tune the model (ranging from 2 to 2). Breiman. Random forests. Machine learning (2001) vol. 45 (l) pp. 5-32.
· Boosted Tree: candidate models had interaction depths raining from 2 to 10, while the number of boosting iterations ranged from 100 to 2000. Friedman. Greedy function approximation: a gradient boosting machine. Annals of Statistics (2001) pp. 1 189-1232.
• Partial Least Squares: the number of PLS components varied from 1 to 20.
• Sparse Partial Least Squares: the number of PLS components varied from 1 to 20 and the regularization parameter ranged from 0.1 to 0.9. Partial Least Squares: Martens and
Naeses, Multivariate calibration. (1989) Wiley; Sparse Partial Least Squares: Chun H, Keles S: Sparse partial least squares for simultaneous dimension reduction and variable selection. Journal of the Royal Statistical Society 2009, 182(l):79-90 • Flexible Discriminant Analysis: candidate models for the MARS basis function used from 2 to 63 terms. Flexible Discriminant Analysis: Hastie et al. Flexible Discriminant Analysis by Optimal Scoring. Journal of the American statistical association (1994) vol. 89 (428).
· Naive Bayes: models were computed using a Gaussian kernel or a non-parametric density estimate.
• Nearest Shrunken Centroids: candidate models used threshold parameters ranging from 0.092 to 2.583. Nearest Shrunken Centroids: Tibshirani et al. Class prediction by nearest shrunken centroids, with applications to DNA microarrays. Statistical Science (2003) pp. 104-117.
• Logistic Regression: weight decay/regularization was used to stabilize the model.
Parameters ranging from 0 to 0.1 were tested. The Elements of Statistical Learning: Data Mining, Inference, and Prediction (Second Edition) by Trevor Hastie, Robert Tibshirani and Jerome Friedman (2009).
· K-Nearest Neighbors: the number of neighbors tested ranged from 5 to 23. The
Elements of Statistical Learning: Data Mining, Inference, and Prediction (Second Edition) by Trevor Hastie, Robert Tibshirani and Jerome Friedman (2009).
• Support Vector Machines: a radial basis function was used with cost parameter values ranging from 0.1 to 100. The Elements of Statistical Learning: Data Mining, Inference, and Prediction (Second Edition) by Trevor Hastie, Robert Tibshirani and Jerome
Friedman (2009).
For each model, the tuning parameter used 50 iterations of the bootstrap to compute resampled estimates of the sensitivity and specificity. Previous modeling for these data showed that high specificity can be easily attained, so the final model was chosen by maximizing the sensitivity value across the candidate models.
Results
Sensitivity and specificity were used to evaluate the data on the training and test sets. On the training set, 50 iterations of the bootstrap were used to get reasonable estimates of performance and to differentiate models. Sensitivity, specificity and the area under the ROC curve were also calculated on the 43 samples in the test set. Table 5 shows the results for the training data across all the models. Table 6 also contains the test set results. Figure 6 shows these results in graphical terms. Table 5: Training set resampling results across various models
:··.:)¾ S;|5«
Boosted I se O.MS e '.7 1 4
FDA 0.537 aS O
K N
logistic ί'ΛνΚ
Figure imgf000064_0001
FLfS 0,201
Jt-iiid' iiii K Ksis 0,76y
Slirnuko GeiilMid-i ') 4 1
Spars PUS itMt) « '.7¾>
SVM (L¾63 ϊ . 67
Table 6: Test set results across various models
mo el A UG
O.ME) 0.80 0.73!
FDA 0.611 o,s¾ 0.764
Figure imgf000064_0002
Logistic Ri.-g 0.611 0,72 0.71»
Naivsj B& is 0.611 I i t
FLS «1667 o <:o
i :. -n l. -ni I¾reste *> ;·.!::· 0.72 ■ 1 il
tn in: l. fi ·' »r I . > 0'. 0.723
:· i: Ρ-> · FLB 0.611 0,72 0.7-19
0.667 0,84 0.7iB
Figure 1 shows the ROC curves for the test set with each model. The solid black point shows the sensitivity/specificity combination that corresponds to using a cutoff of 0.50 on the probability estimate of being Alzheimer's disease. The solid square indicates an alternate choice based on getting the sensitivity as close as possible. The test in each panel shows the alternative cutoff and the corresponding sensitivity/specificity estimates.
Without wishing to be bound by any particular theory, it is believed that caution should be taken when interpreting the results from the alternate cutoff. Since ROC curve analysis is part of the modeling process, any new cutoff should be evaluated on a separate data set.
Figure 7 also shows the sensitivity and specificity profiles across various probability cutoffs. This plot can demonstrate how adaptable each model is to alternative cutoffs. It was observed from the models that were run on these data, the partial least squares (PLS) model appeared to have the best combination of sensitivity and specificity, as well as a high test set ROC AUC value. The PLS model used all the predictors in the data set.
Table 7 has a list of the top 15 predictors that contribute to the model. The overall distribution of the scores is shown in Figure 8. In Figure 8, there is not clear cluster of variables with "high" and "low" importance. The rankings of the genotype variables were not in the top 15.
Table 7: The top 15 most important predictors for the PLS model (from most important to least)
Overall
ΓΊ:ν·
(m
Figure imgf000065_0001
-■U-i i Hi.'
ApoD ! IIS
CLCSF if.011;
;p :.b-n., c \'<<;
A; >,L !i.0100
··:,·<: Λ VI if.OlOO
UMP2 Oijij!!iiO
FDGP_BB 'i,0OE. i
S'.U'jiiJO
iM'P!
t_3(¾
For the top 15, Figure 9 shows the correlation patterns between the variables and Figure 8 shows the distribution of the training set data for each predictor. Figure 10 shows no single predictor that significantly differentiates Alzheimer's disease from control. This also supports the idea that the models require a large number of predictors to achieve adequate performance.
Table 8 and 9 is based on the univariate Analysis of Covariance
(ANCOVA) models comparing the two phenotypes (AD vs other) while adjusting also for age and gender of the subjects. The p-values supplied are based on the type III sums of squares, thereby reflecting the ability of the marker (on its own, not in conjunction with other markers) to distinguish between the two groups above and beyond the ability of age and gender to discriminate. Table 8: UPenn Plasma Univariate: Comparison Alzheimer's disease vs Control only
Response Log Alpha 1 -Antichymotrypsin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.09757082 4.9286 0.0275
Age 1 1 0.00632811 0.3197 0.5724
Gender 1 1 0.02083983 1.0527 0.3060
Response Log Alpha-1 Antitrypsin Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.11654156 12.7513 0.0004
Age 1 1 0.00296641 0.3246 0.5695
Gender 1 1 0.01436133 1.5713 0.2114
Response Log ACE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00893135 0.3298 0.5664
Age 1 1 0.06297906 2.3254 0.1288
Gender 1 1 0.13529560 4.9955 0.0264
Response Log Adiponectin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.10072464 1.4883 0.2238
Age 1 1 0.20830292 3.0778 0.0808
Gender 1 1 0.95091021 14.0503 0.0002
Response Log Agouti Related Protein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.05191313 0.6177 0.4328
Age 1 1 0.22554090 2.6838 0.1028
Gender 1 1 0.14469766 1.7218 0.1909
Response Log Alpha-2 Macroglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.01371995 2.6884 0.1025
Age 1 1 0.00418567 0.8202 0.3661
Gender 1 1 0.10168754 19.9256 <.0001
Response Log Alpha-Fetoprotein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0039838 0.0323 0.8575
Age 1 1 1.6238383 13.1662 0.0004
Gender 1 1 0.8761198 7.1036 0.0083
Response Log Alphal Microglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.03956080 2.3233 0.1289
Age 1 1 0.19675816 11.5548 0.0008
Gender 1 1 0.02769081 1.6262 0.2036
Response Log ANG-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00742880 0.2975 0.5860
Age 1 1 0.99784496 39.9622 <.0001
Gender 1 1 0.07306225 2.9260 0.0886
Response Log Angiotensinogen Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0691295 0.0879 0.7672
Age 1 1 1.2130149 1.5418 0.2157
Gender 1 1 1.5113540 1.9210 0.1672
Response Log Apo A1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.13548238 5.7580 0.0173
Age 1 1 0.00010260 0.0044 0.9474
Gender 1 1 0.46759744 19.8730 <.0001
Response Log Apo A2 Effect Tests
Source Nparm Sum of Squares F Ratio Prob > F
DIAG 1 0.00150715 0.1137 0.7363
Age 1 0.02247936 1.6956 0.1943 Source N arm DF Sum of Squares F Ratio Prob > F Gender 1 1 0.09984367 7.5313 0.0066
Response Log ApoCI Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00001020 0.0008
Age 1 1 0.02724053 2.0179
Gender 1 1 0.42493871 314785
Response Log Apo Clll Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00055256 0.0244
Age 1 1 0.00168450 0.0744
Gender 1 1 0.27464797 12.1233
Response Log ApoH Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.10012685 8.1323
Age 1 1 0.00380708 0.3092
Gender 1 1 0.00001769 0.0014
Response Log Apo A-IV Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00032618 0.0078 0.9297 Age 1 1 0.06832497 1.6356 0.2023 Gender 1 1 0.08021122 1.9201 0.1673 Response Log ApoB Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02689237 2.2022
Age 1 1 0.08758695 7.1725
Gender 1 1 0.04288546 3.5119
Response Log ApoD Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.10001944 5.6304
Age 1 1 0.00897956 0.5055
Gender 1 1 0.22521563 12.6780
Response Log ApoE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.20639489 6.5366 0.0113
Age 1 1 0.02060278 0.6525 0.4201
Gender 1 1 0.63626015 20.1505 <.0001
Response Log ApoJ Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00214321 0.4480 0.5040
Age 1 1 0.01316376 2.7515 0.0986
Gender 1 1 0.08331881 17.4154 <.0001
Response Log AXL Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00470212 0.2133 0.6446
Age 1 1 0.00018707 0.0085 0.9267
Gender 1 1 0.03260202 1.4791 0.2252
Response Log Beta2 Microglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04007431 2.9127 0.0893 Age 1 1 0.66365011 48.2355 <.0001 Gender 1 1 0.00331402 0.2409 0.6241
Response Log BLC Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.04064608 0.5990
Age 1 1 0.65398649 9.6370
Gender 1 1 0.03176943 0.4681
Response Log BMP -6 Effect Tests
Source Nparm Sum of Squares F Ratio Prob > F DIAG 1 0.01049858 0.1622 0.6875 Source N arm DF Sum of Squares F Ratio Prob > F
Age 1 1 0.30090814 4.6502 0.0322
Gender 1 1 0.26789654 4.1401 0.0431
Response Log BDNF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.9739714 3.9635
Age 1 1 2.3079839 9.3922
Gender 1 1 0.0059720 0.0243
Response Log Betacellulin Effect Tests
Source N arm DF Sum of Squares F Ratio
DIAG 1 1 0.27314883 3.1594
Age 1 1 0.04122411 0.4768
Gender 1 1 0.04713463 0.5452
Response Clean C3 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03250361 1.5681
Age 1 1 0.05851025 2.8228
Gender 1 1 0.19054226 9.1927
Response Log Cancer Antigen 19-9 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0104371 0.0217 0.8830 Age 1 1 1.0640636 2.2148 0.1382 Gender 1 1 0.0913984 0.1902 0.6632
Response Log CD40 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.11245269 5.5735 0.0191 Age 1 1 0.39083707 19.3712 <.0001 Gender 1 1 0.00015022 0.0074 0.9313
Response Log CD40 Ligand Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.3263141 3.4310
Age 1 1 1.2308276 12.9415
Gender 1 1 0.0092553 0.0973
Response Log CDL5 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.06907743 2.6644 0.1041
Age 1 1 0.33110385 12.7710 0.0004
Gender 1 1 0.00551303 0.2126 0.6452
Response Log Carcinoembryonic Antigen Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0268749 0.3593 0.5495 Age 1 1 1.3395152 17.9082 <.0001 Gender 1 1 0.0015312 0.0205 0.8864
Response Log CgA Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.0153585 0.2311
Age 1 1 2.4438349 36.7693
Gender 1 1 0.0002601 0.0039
Response Log CKMB Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.29278936 4.5918
Age 1 1 0.51286060 8.0431
Gender 1 1 0.70203279 11.0099
Response Log CNTF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.09135214 1.1767
Age 1 1 0.09781011 1.2599
Gender 1 1 0.09022008 1.1621
Response Log Complement Factor H Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00474600 0.1504 0.6985 Age 1 1 0.04241890 1.3445 0.2475 Gender 1 1 0.00213276 0.0676 0.7951
Response Log CTGF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.08293324 2.1646
Age 1 1 0.00741676 0.1936
Gender 1 1 0.23071753 6.0218
Response Log Cortisol Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.22555210 13.3566
Age 1 1 0.00430777 0.2551
Gender 1 1 0.10357875 6.1337
Response Log C-Peptide Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00075280 0.0078
Age 1 1 0.81716606 84666
Gender 1 1 0.62064905 64305
Response Log CRP Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 1.4184758 4.6475
Age 1 1 0.2229228 0.7304
Gender 1 1 0.9091701 2.9788
Response Log Cystatin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06347550 4.2353
Age 1 1 0.32585564 21.7424
Gender 1 1 0.03909462 2.6086
Response Log EGF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.6643216 5.4834
Age 1 1 1.4071244 11.6147
Gender 1 1 0.0393685 0.3250
Response Log EGF-R Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.04021177 1.8645 0.1735
Age 1 1 0.20639230 9.5699 0.0022
Gender 1 1 0.00653961 0.3032 0.5824
Response Log ENA-78 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.28983488 1.9380 0.1653
Age 1 1 0.26394974 1.7649 0.1854
Gender 1 1 0.33790605 2.2594 0.1343
Response Log Endothelin-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00518124 0.0836
Age 1 1 0.01042439 0.1682
Gender 1 1 0.09779083 1.5782
Response Log EN-RAGE Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.48257884 6.4912
Age 1 1 0.12175491 1.6377
Gender 1 1 0.16606285 2.2337
Response Log Eotaxin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00089570 0.0099
Age 1 1 0.13198730 1.4561
Gender 1 1 0.04421998 0.4878 Response Log E-Selectin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.14921791 4.1136
Age 1 1 0.00233491 0.0644
Gender 1 1 0.01617998 0.4461
Response Log FABP Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.0136922 0.0673
Age 1 1 6.2144717 30.5362
Gender 1 1 1.2982004 6.3790
Response Clean Factor VII Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 7896.37 0.2188 0.6404
Age 1 1 3266.43 0.0905 0.7638
Gender 1 1 606208.80 16.7959 <.0001
Response Log FAS Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.05636966 3.8801
Age 1 1 0.15077612 10.3784
Gender 1 1 0.10987253 7.5629
Response Log Fas-Ligand Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.02145776 0.4647 0.4962 Age 1 1 0.22218847 4.8122 0.0293 Gender 1 1 0.15718221 3.4043 0.0664
Response Log Ferritin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0401446 0.2679 0.6053 Age 1 1 0.5584810 3.7264 0.0549 Gender 1 1 1.4988784 10.0012 0.0018
Response Log Fetuin A Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00513336 0.8342
Age 1 1 0.07751276 12.5969
Gender 1 1 0.02472305 4.0179
Response Log Fibrinogen Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06487349 4.1894
Age 1 1 0.00689806 0.4455
Gender 1 1 0.06763528 4.3678
Response Log FSH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0022382 0.0650 0.7991
Age 1 1 0.2907121 8.4361 0.0041
Gender 1 1 6.3655922 184.7220 <.0001
Response Log G-CSF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.33721686 8.3831 0.0042
Age 1 1 0.04529264 1.1260 0.2898
Gender 1 1 0.27247284 6.7736 0.0099
Response Log Growth Hormone Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.1319637 0.3275 0.5677
Age 1 1 4.3569639 10.8132 0.0012
Gender 1 1 1.8833330 4.6741 0.0317
Response Log GLP-1 Total Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00196054 0.0675 0.7953
Age 1 1 0.00007370 0.0025 0.9599
Gender 1 1 0.07023159 2.4168 0.1215 Response Log Glucagon Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06088236 1.5376
Age 1 1 0.05101071 1.2883
Gender 1 1 0.11944776 3.0166
Response Log GST alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.51211036 3.7203
Age 1 1 0.30192565 2.1934
Gender 1 1 0.51540279 3.7442
Response Log GRO-alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.39391010 4.0260
Age 1 1 0.05061264 0.5173
Gender 1 1 0.00258595 0.0264
Response Log Haptoglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.20300486 0.7516
Age 1 1 0.04462344 0.1652
Gender 1 1 0.58762986 2.1757
Response Log HB-EGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.1454743 0.7115 0.3999
Age 1 1 1.1304977 5.5290 0.0196
Gender 1 1 0.4959013 2.4253 0.1209
Response Log HCC-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.03513513 0.7691 0.3815
Age 1 1 0.12077438 2.6436 0.1054
Gender 1 1 0.01826546 0.3998 0.5279
Response Log HGF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00449682 0.2871
Age 1 1 0.03281238 2.0953
Gender 1 1 0.06866183 4.3845
Response Log HSP60 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.21681869 2.5026
Age 1 1 0.02382186 0.2750
Gender 1 1 0.53678302 6.1957
Response Log I-309 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 1.0215429 4.4009
Age 1 1 0.1730087 0.7453
Gender 1 1 0.0013619 0.0059
Response Log ICAM-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00169748 0.0885 0.7664
Age 1 1 0.04991320 2.6011 0.1083
Gender 1 1 0.08234349 4.2912 0.0395
Response Log IgA Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.24489265 5.9353 0.0157
Age 1 1 0.01852248 0.4489 0.5036
Gender 1 1 0.00208025 0.0504 0.8226
Response Log IgE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.3139331 0.6930 0.4061
Age 1 1 0.7377087 1.6285 0.2033
Gender 1 1 2.5200239 5.5630 0.0193 Response Log IGFBP-2 Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.6420886 10.7303 0.0012 Age 1 1 2.2682407 37.9059 <.0001 Gender 1 1 0.0006157 0.0103 0.9193
Response Log IgM Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00878421 0.1284
Age 1 1 0.08182886 1.1958
Gender 1 1 0.07404381 1.0820
Response Log IL-10 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.10788428 3.6404
Age 1 1 0.14722877 4.9680
Gender 1 1 0.00493087 0.1664
Response Log IL-12p40 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.31524616 7.9071 0.0054 Age 1 1 0.02984605 0.7486 0.3879 Gender 1 1 0.00668055 0.1676 0.6827
Response Log IL-13 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.17723383 4.0742 0.0448 Age 1 1 0.08598180 1.9765 0.1612 Gender 1 1 0.00656014 0.1508 0.6982
Response Log IL-15 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.12650420 2.6982 0.1019 Age 1 1 0.00454178 0.0969 0.7559 Gender 1 1 0.00174762 0.0373 0.8471
Response Log IL-16 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02167414 1.1274
Age 1 1 0.14040023 7.3031
Gender 1 1 0.01948536 1.0136
Response Log IL-18 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00000090 0.0000
Age 1 1 0.02999650 0.9353
Gender 1 1 0.18201466 5.6754
Response Log IL-1 ra Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00459148 0.0856
Age 1 1 0.00000672 0.0001
Gender 1 1 0.25180684 4.6921
Response Log IL-3 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.30101880 4.2495
Age 1 1 0.11602487 1.6379
Gender 1 1 0.01103847 0.1558
Response Log IL-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.11663309 2.9494
Age 1 1 0.00768277 0.1943
Gender 1 1 0.00092567 0.0234
Response Log IL-5 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.11630491 2.8059 0.0954
Age 1 1 0.00001771 0.0004 0.9835
Gender 1 1 0.08699301 2.0987 0.1489 Response Log IL-6 Receptor Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00000365 0.0002
Age 1 1 0.00745633 04033
Gender 1 1 0.00047136 0.0255
Response Log IL-7 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.15411911 1.8845 0.1713 Age 1 1 0.22255314 2.7213 0.1005 Gender 1 1 0.00744424 0.0910 0.7632
Response Log IL-8 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04554019 1.9240 0.1669 Age 1 1 0.64728321 27.3474 <.0001 Gender 1 1 0.00614616 0.2597 0.6109
Response Log Insulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03248195 0.2073 0.6493 Age 1 1 0.33195516 2.1190 0.1469 Gender 1 1 0.29256644 1.8676 0.1732
Response Log IP-10 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0032570 0.1000 0.7521 Age 1 1 1.2522396 38.4663 <.0001 Gender 1 1 0.1343761 4.1278 0.0434
Response Log Kidney Injury Molecule Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02039153 0.2132 0.6447
Age 1 1 0.05990714 0.6263 0.4296
Gender 1 1 0.01620316 0.1694 0.6810
Response Log Leptin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.1314386 0.8608 0.3546 Age 1 1 0.0101785 0.0667 0.7965 Gender 1 1 9.0073937 58.9869 <.0001
Response Log LH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.02833032 0.5889 0.4437 Age 1 1 0.01620949 0.3369 0.5622 Gender 1 1 0.75698779 15.7349 <.0001
Response Log Lipoprotein (a) Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.1810974 0.5969
Age 1 1 1.1556530 3.8091
Gender 1 1 0.8659218 2.8541
Response Log MCP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00477385 0.1678 0.6825
Age 1 1 0.00796370 0.2800 0.5973
Gender 1 1 0.12038512 4.2324 0.0409
Response Log MCP-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.06396664 1.9093 0.1685
Age 1 1 0.13004599 3.8816 0.0501
Gender 1 1 0.00159426 0.0476 0.8275
Response Log MCP-3 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.08305116 0.4915
Age 1 1 0.14898399 0.8817
Gender 1 1 0.01579239 0.0935 Response Log MCP-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.35626731 2.8787 0.0912
Age 1 1 0.22240254 1.7971 0.1815
Gender 1 1 0.07504485 0.6064 0.4370
Response Log M-CSF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00343069 0.5612 0.4546
Age 1 1 0.01211031 1.9812 0.1607
Gender 1 1 0.00403273 0.6597 0.4176
Response Log MDC Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.11441224 4.3831 0.0375
Age 1 1 0.02455237 0.9406 0.3332
Gender 1 1 0.14380126 5.5089 0.0198
Response Log MIF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00245775 0.0354 0.8509 Age 1 1 0.00015384 0.0022 0.9625 Gender 1 1 0.09223797 1.3292 0.2502
Response Log Gamma Interferon Induced Monokine Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.1635090 2.2649 0.1338 Age 1 1 6.2141380 86.0754 <.0001 Gender 1 1 0.0233275 0.3231 0.5703
Response Log MIP-1 alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01490893 0.7751 0.3796 Age 1 1 0.35613664 18.5141 <.0001 Gender 1 1 0.00430177 0.2236 0.6368
Response Log MIP-1 beta Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.16751430 5.6689 0.0181 Age 1 1 0.28575218 9.6703 0.0021 Gender 1 1 0.00428598 0.1450 0.7037
Response Log MIP-3 alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01761420 0.3777 0.5395 Age 1 1 0.08578056 1.8392 0.1765 Gender 1 1 0.00882108 0.1891 0.6641
Response Log MMP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.09167418 1.8845 0.1713 Age 1 1 0.12919087 2.6558 0.1046 Gender 1 1 0.00451312 0.0928 0.7610
Response Log MMP-10 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.04351991 2.2109 0.1385
Age 1 1 0.00903112 0.4588 0.4989
Gender 1 1 0.07561340 3.8413 0.0513
Response Log MMP-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01970485 1.7855 0.1829 Age 1 1 0.52235119 47.3301 <.0001 Gender 1 1 0.00027807 0.0252 0.8740
Response Log MMP-7 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.49766396 8.2502 0.0045 Age 1 1 0.45035758 7.4660 0.0068 Gender 1 1 0.05850088 0.9698 0.3258 Response Log MMP9 (total) Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.01492707 0.3738 0.5416
Age 1 1 0.03414682 0.8550 0.3562
Gender 1 1 0.00124679 0.0312 0.8599
Response Log MMP-9 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00556767 0.1064 0.7446
Age 1 1 0.02464506 04710 0.4933
Gender 1 1 0.00672152 0.1285 0.7204
Response Log Myeloid Progenitor Inhibitory Factor 1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00169608 0.0457 0.8310
Age 1 1 0.02595498 0.6991 0.4040
Gender 1 1 0.03353098 0.9032 0.3430
Response Log Myeloperoxidase Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.10312558 2.7195 0.1006
Age 1 1 0.03081056 0.8125 0.3684
Gender 1 1 0.14757376 3.8917 0.0498
Response Log Myoglobin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0128826 0.2308 0.6314
Age 1 1 0.9620369 17.2382 <.0001
Gender 1 1 1.5553831 27.8700 <.0001
Response Log NGAL Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.10265722 4.9015 0.0279
Age 1 1 0.12790852 6.1071 0.0142
Gender 1 1 0.01304547 0.6229 0.4309
Response Log NrCAM Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02501126 0.8593 0.3550
Age 1 1 0.07681880 2.6393 0.1057
Gender 1 1 0.01927987 0.6624 0.4166
Response Log NT-proBNP Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.3294675 2.7814 0.0968
Age 1 1 7.5929940 64.1001 <.0001
Gender 1 1 0.4931799 4.1634 0.0425
Response Log Osteopontin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.69657598 11.3035 0.0009
Age 1 1 0.41519074 6.7374 0.0101
Gender 1 1 0.00798441 0.1296 0.7192
Response Log PAI-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.84859772 7.8749 0.0055
Age 1 1 0.44954157 4.1717 0.0423
Gender 1 1 0.08725773 0.8097 0.3692
Response Log Pancreatic Polypeptide Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.5332216 4.9389 0.0273
Age 1 1 1.1143541 10.3216 0.0015
Gender 1 1 0.4182280 3.8738 0.0503
Response Log Prostatic Acid Phosphatase Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0070432 0.1998 0.6554
Age 1 1 0.0239748 0.6800 0.4105
Gender 1 1 1.7600504 49.9197 <.0001 Response Log PAPP-A Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.02941113 0.9495 0.3309 Age 1 1 0.15681323 5.0628 0.0255 Gender 1 1 0.39764860 12.8382 0.0004
Response Log PARC Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00218937 0.2004 0.6548 Age 1 1 0.00189119 0.1731 0.6778 Gender 1 1 0.00123846 0.1134 0.7367
Response Log PDGF-BB Effect Tests
Source N arm DF Sum of Squares F Ratio
DIAG 1 1 2.1888090 12.0703
Age 1 1 0.6790357 3.7446
Gender 1 1 0.1269765 0.7002
Response Log PLGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.02038334 0.2180 0.6410 Age 1 1 0.05618906 0.6010 0.4390 Gender 1 1 0.42403864 4.5359 0.0343
Response Log Progesterone Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.1339600 1.4964 0.2226 Age 1 1 1.0648380 11.8946 0.0007 Gender 1 1 0.4020581 4.4911 0.0352
Response Log Proinsulin, Intact Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0496975 0.4180 0.5186
Age 1 1 1.3717368 11.5379 0.0008
Gender 1 1 0.5645608 4.7486 0.0304
Response Log Proinsulin, Total Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.0041617 0.0368
Age 1 1 1.5782284 13.9460
Gender 1 1 0.4999796 4.4181
Response Log Prolactin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06380200 2.8963
Age 1 1 0.00159471 0.0724
Gender 1 1 0.07836238 3.5573
Response Clean Protein S Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 43.468069 3.6411
Age 1 1 25.415184 2.1289
Gender 1 1 5.438644 0.4556
Response Log PYY Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.17971507 2.0659 0.1521 Age 1 1 0.61364942 7.0541 0.0085 Gender 1 1 0.00082324 0.0095 0.9226
Response Log RANTES Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 1.0496024 5.3433 0.0218
Age 1 1 1.3382554 6.8128 0.0097
Gender 1 1 0.1576702 0.8027 0.3713
Response Log Resistin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.27339616 10.7565 0.0012
Age 1 1 0.19935100 7.8432 0.0056
Gender 1 1 0.00751942 0.2958 0.5871 Response Clean Serum Amyloid P Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 106.04379 5.4764 0.0202
Age 1 1 1.80531 0.0932 0.7604
Gender 1 1 87.55513 4.5216 0.0346
Response Log Stem Cell Factor Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.01852026 1.1148
Age 1 1 0.32018466 19.2730
Gender 1 1 0.01426403 0.8586
Response Log SGOT Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04847391 4.0115 0.0465 Age 1 1 0.02163685 1.7906 0.1823 Gender 1 1 0.00009816 0.0081 0.9283
Response Log SHBG Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.4339200 10.6601 0.0013 Age 1 1 0.5003729 12.2926 0.0006 Gender 1 1 1.2732352 31.2795 <.0001
Response Log SOD Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00132145 0.0321 0.8579 Age 1 1 0.06223609 1.5132 0.2200 Gender 1 1 0.01496311 0.3638 0.5470
Response Log Sortilin
Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04247027 4.6459 0.0322 Age 1 1 0.17688474 19.3498 <.0001 Gender 1 1 0.05852244 6.4019 0.0121
Response Log sRAGE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.22861903 3.3367 0.0691 Age 1 1 0.00000033 0.0000 0.9982 Gender 1 1 0.07176324 1.0474 0.3073
Response Log Tamm-Horsfall Protein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00539457 0.1134 0.7367
Age 1 1 0.44474727 9.3467 0.0025
Gender 1 1 0.53750842 11.2962 0.0009
Response Log Thyroxine Binding Globulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01656804 2.4247 0.1209 Age 1 1 0.00211486 0.3095 0.5786 Gender 1 1 0.09915053 14.5107 0.0002 Response Log TECK
Source Nparm Sum of Squares F Ratio Prob > F DIAG 1 0.00203065 0.0743 0.7854 Age 1 0.04045441 1.4807 0.2250 Gender 1 0.00243171 0.0890 0.7657
Response Log Tenasci Effect Tests
Source Nparm Sum of Squares F Ratio
DIAG 1 0.54092106 8.3190
Age 1 0.00836436 0.1286
Gender 1 0.06827918 1.0501
Response Log Testosterone Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.002196 0.0688 0.7933
Age 1 1 0.688003 21.5645 <.0001
Gender 1 1 12.640627 396.2021 <.0001 Response Log TGF-alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.12985650 1.1246
Age 1 1 0.00027109 0.0023
Gender 1 1 0.57510231 4.9805
Response Log Thrombomodulin Effect Tests
Source N arm DF Sum of Squares F Ratio
DIAG 1 1 0.11910394 7.4531
Age 1 1 0.22303312 13.9566
Gender 1 1 0.08343555 5.2211
Response Log TIMP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00448977 0.3429 0.5588 Age 1 1 0.31317984 23.9173 <.0001 Gender 1 1 0.05852278 4.4693 0.0357
Response Log TNFRII Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.05342657 2.3887 0.1237 Age 1 1 0.86126313 38.5072 <.0001 Gender 1 1 0.05332364 2.3841 0.1241
Response Log Thrombopoietin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.01263842 0.5889
Age 1 1 0.06204650 2.8909
Gender 1 1 0.01252604 0.5836
Response Log TRAIL R3 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.01891152 0.5502
Age 1 1 0.03540012 1.0299
Gender 1 1 0.01459360 0.4246
Response Clean Transferrin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 36596.45 0.3276 0.5677 Age 1 1 651439.43 5.8307 0.0166 Gender 1 1 618910.26 5.5395 0.0195
Response Log Trefoil Factor 3 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0515857 1.5963 0.2078 Age 1 1 1.1693789 36.1866 <.0001 Gender 1 1 0.1108203 3.4293 0.0654
Response Log TSH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03925779 0.6584 0.4180 Age 1 1 0.00408833 0.0686 0.7937 Gender 1 1 0.01214189 0.2036 0.6523
Response Log Thrombospondin-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 1.0240591 3.9154 0.0491 Age 1 1 2.0486098 7.8327 0.0056 Gender 1 1 0.0075214 0.0288 0.8655
Response Clean TTR Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 1575.074 0.5561 0.4567 Age 1 1 10815.801 3.8187 0.0520 Gender 1 1 42562.481 15.0274 0.0001
Response Log VCAM Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03325608 2.5553 0.1114 Age 1 1 0.23542032 18.0892 <.0001 Gender 1 1 0.00586458 0.4506 0.5028 Response Log VEGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.07248942 2.8019 0.0956
Age 1 1 0.17934262 6.9321 0.0091
Gender 1 1 0.04050143 1.5655 0.2122
Response Clean Vitronectin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 99.155 0.0093 0.9232
Age 1 1 16652.525 1.5630 0.2126
Gender 1 1 6980.347 0.6552 0.4192
Response Log von Willebrand Factor Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.05169529 0.8901 0.3465
Age 1 1 0.55639546 9.5799 0.0022
Gender 1 1 0.00094014 0.0162 0.8989
Table 9: UPenn Plasma Alzheimer's disease vs Other Univariate. Age and Gender
Response Log Alpha 1 -Antichymotrypsin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00309601 0.1352 0.7137
Age 1 1 0.01101623 04810 0.4892
Gender 1 1 0.02913031 1.2719 0.2614
Response Log Alpha-1 Antitrypsin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00003468 0.0049 0.9441
Age 1 1 0.00426070 0.6066 0.4374
Gender 1 1 0.02097812 2.9866 0.0862
Response Log ACE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.12788902 4.3604 0.0386
Age 1 1 0.01613932 0.5503 0.4595
Gender 1 1 0.00000121 0.0000 0.9949
Response Log Adiponectin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.10341838 1.6151 0.2059
Age 1 1 0.14212153 2.2196 0.1386
Gender 1 1 045887861 7.1666 0.0083
Response Log Agouti Related Protein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.03304685 04656 04962
Age 1 1 0.28697202 4.0428 0.0463
Gender 1 1 0.66364604 9.3492 0.0027
Response Log Alpha-2 Macroglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.04419461 9.2495 0.0028
Age 1 1 0.00086357 0.1807 0.6714
Gender 1 1 0.02327618 4.8715 0.0290
Response Log Alpha-Fetoprotein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0089983 0.1033 0.7484
Age 1 1 1.5809929 18.1484 <.0001
Gender 1 1 1.0699383 12.2820 0.0006
Response Log Alphal Microglobulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00008132 0.0042 0.9484 Age 1 1 0.06719291 34673 0.0648 Gender 1 1 0.00355048 0.1832 0.6693
Response Log ANG-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06247723 2.5246
Age 1 1 0.20147025 8.1412
Gender 1 1 0.05364905 2.1679
Response Log Angiotensinogen Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00390611 0.0053 0.9419 Age 1 1 0.17683485 0.2416 0.6239 Gender 1 1 0.02410816 0.0329 0.8563
Response Log Apo A1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.35398992 14.3085
Age 1 1 0.00227715 0.0920
Gender 1 1 0.23558010 9.5223
Response Log Apo A2 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00054261 0.0457
Age 1 1 0.01237080 1.0419
Gender 1 1 0.09003143 7.5823
Response Log ApoCI Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00457215 0.3685
Age 1 1 0.00234256 0.1888
Gender 1 1 0.14493017 11.6798
Response Log Apo Clll Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03182938 1.4841
Age 1 1 0.05736538 2.6748
Gender 1 1 0.52922429 24.6765
Response Log ApoH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00000022 0.0000 0.9959 Age 1 1 0.00163374 0.1942 0.6601 Gender 1 1 0.00151648 0.1803 0.6718
Response Log Apo A-IV Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01198971 0.2649 0.6076 Age 1 1 0.01298367 0.2869 0.5931 Gender 1 1 0.00250276 0.0553 0.8144
Response Log ApoB Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00747490 0.5144
Age 1 1 0.01068622 0.7353
Gender 1 1 0.03065425 2.1094
Response Log ApoD Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.27842576 15.3916 0.0001
Age 1 1 0.00140484 0.0777 0.7809
Gender 1 1 0.29054201 16.0614 0.0001
Response Log ApoE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.15412082 4.3330 0.0393
Age 1 1 0.01895254 0.5328 0.4667
Gender 1 1 0.32049486 9.0106 0.0032 Response Log ApoJ Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00076333 0.1500
Age 1 1 0.00105723 0.2077
Gender 1 1 0.07243679 14.2298
Response Log AXL Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06408687 3.1370
Age 1 1 0.00042900 0.0210
Gender 1 1 0.00318474 0.1559
Response Log Beta2 Microglobulin Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00018430 0.0111 0.9164 Age 1 1 0.21665283 12.9922 0.0004 Gender 1 1 0.00228699 0.1371 0.7117
Response Log BLC Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.13284888 2.0349
Age 1 1 0.19277850 2.9528
Gender 1 1 0.00007850 0.0012
Response Log BMP -6 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.01137700 0.1279 0.7212
Age 1 1 0.40209494 4.5194 0.0353
Gender 1 1 0.05096163 0.5728 0.4505
Response Log BDNF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 5.8863603 26.9745 <.0001
Age 1 1 1.2370992 5.6691 0.0187
Gender 1 1 0.1285341 0.5890 0.4441
Response Log Betacellulin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.44644167 54089
Age 1 1 0.00446691 0.0541
Gender 1 1 0.24531432 2.9721
Response Clean C3 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.07107676 3.1372
Age 1 1 0.00130196 0.0575
Gender 1 1 0.19615605 8.6579
Response Log Cancer Antigen 19-9 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00010009 0.0002 0.9883 Age 1 1 0.14507591 0.3135 0.5765 Gender 1 1 0.31673344 0.6844 0.4095
Response Log CD40 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00067406 0.0311 0.8602
Age 1 1 0.07099809 3.2786 0.0724
Gender 1 1 0.02694844 1.2444 0.2666
Response Log CD40 Ligand Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 5.3333244 45.8487 <.0001 Age 1 1 0.8491355 7.2997 0.0078 Gender 1 1 0.0041466 0.0356 0.8505
Response Log CDL5 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.07212531 2.0438 0.1551
Age 1 1 0.18976651 5.3773 0.0219
Gender 1 1 0.02635455 0.7468 0.3890 Response Log Carcinoembryonic Antigen Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.2552224 3.8234 0.0526 Age 1 1 1.9304848 28.9197 <.0001 Gender 1 1 0.0010399 0.0156 0.9009
Response Log CgA Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03239612 0.5005
Age 1 1 0.99289559 15.3401
Gender 1 1 0.01869886 0.2889
Response Log CKMB Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.2927776 4.9816
Age 1 1 0.3491461 5.9407
Gender 1 1 1.0315442 17.5517
Response Log CNTF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.08330005 1.1829
Age 1 1 0.00113542 0.0161
Gender 1 1 0.00605224 0.0859
Response Log Complement Factor H Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00430038 0.1146 0.7355 Age 1 1 0.00018750 0.0050 0.9437 Gender 1 1 0.02437967 0.6498 0.4216
Response Log CTGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00043029 0.0083 0.9274 Age 1 1 0.04533892 0.8770 0.3507 Gender 1 1 0.00233893 0.0452 0.8319
Response Log Cortisol Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03407374 2.3088
Age 1 1 0.00588324 0.3986
Gender 1 1 0.06453539 4.3729
Response Log C-Peptide Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.40096318 5.2768
Age 1 1 0.47157244 6.2061
Gender 1 1 0.52455378 6.9033
Response Log CRP Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.69684447 2.1059
Age 1 1 0.17892658 0.5407
Gender 1 1 0.95170846 2.8761
Response Log Cystatin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00149139 0.1000 0.7524
Age 1 1 0.08452885 5.6661 0.0187
Gender 1 1 0.00387165 0.2595 0.6113
Response Log EGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 5.7831540 43.1596 <.0001
Age 1 1 1.2021661 8.9717 0.0033
Gender 1 1 0.0127866 0.0954 0.7579
Response Log EGF-R Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.07862613 4.6074
Age 1 1 0.01194499 0.7000
Gender 1 1 0.00419388 0.2458 Response Log ENA-78 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 3.5473235 27.0031
Age 1 1 0.8711928 6.6317
Gender 1 1 0.0426209 0.3244
Response Log Endothelin-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02088391 04172
Age 1 1 0.00802419 0.1603
Gender 1 1 0.61337540 12.2523
Response Log EN-RAGE Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00280438 0.0287
Age 1 1 0.00853931 0.0873
Gender 1 1 0.02036726 0.2082
Response Log Eotaxin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.38195213 5.3347
Age 1 1 0.13505149 1.8863
Gender 1 1 0.00712569 0.0995
Response Log E-Selectin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01933584 0.4714 0.4935 Age 1 1 0.00225549 0.0550 0.8150 Gender 1 1 0.01286026 0.3135 0.5764
Response Log FABP Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0155896 0.0720 0.7889 Age 1 1 1.0997873 5.0796 0.0258 Gender 1 1 1.1925547 5.5080 0.0204
Response Clean Factor VII Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 29923.76 0.7733 0.3808
Age 1 1 2673.44 0.0691 0.7931
Gender 1 1 640558.83 16.5528 <.0001 Response Log FAS Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02418242 1.5772
Age 1 1 0.12992281 8.4735
Gender 1 1 0.09010678 5.8767
Response Log Fas-Ligand Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.04207548 1.0085
Age 1 1 0.13578602 3.2545
Gender 1 1 0.00454046 0.1088
Response Log Ferritin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.05577516 0.3341 0.5642
Age 1 1 0.28652951 1.7164 0.1924
Gender 1 1 0.94816993 5.6798 0.0185
Response Log Fetuin A Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00004587 0.0062 0.9373
Age 1 1 0.00173568 0.2353 0.6284
Gender 1 1 0.04963110 6.7278 0.0105
Response Log Fibrinogen Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00096977 0.0680
Age 1 1 0.00082767 0.0581
Gender 1 1 0.04520213 3.1707 Response Log FSH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0180488 04863 0.4868
Age 1 1 0.2073365 5.5865 0.0195
Gender 1 1 4.7420435 127.7710 <.0001
Response Log G-CSF Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00196731 0.0459
Age 1 1 0.00062115 0.0145
Gender 1 1 0.30788138 7.1823
Response Log Growth Hormone Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02155816 0.0457
Age 1 1 0.51065071 1.0830
Gender 1 1 0.60024839 1.2730
Response Log GLP-1 Total Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02058446 0.8561
Age 1 1 0.00673840 0.2803
Gender 1 1 0.15567323 6.4744
Response Log Glucagon Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.04768945 1.2788 0.2601
Age 1 1 0.11411257 3.0599 0.0825
Gender 1 1 0.12646904 3.3912 0.0677
Response Log GST alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.27099280 1.7714 0.1854
Age 1 1 0.02735270 0.1788 0.6731
Gender 1 1 0.22099099 14446 0.2315
Response Log GRO-alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 2.7151783 30.4431
Age 1 1 0.2560178 2.8705
Gender 1 1 0.0549920 0.6166
Response Log Haptoglobul Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.98291792 3.1142
Age 1 1 0.04589758 0.1454
Gender 1 1 0.28813856 0.9129
Response Log HB-EGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 3.6513564 17.7594 <.0001 Age 1 1 0.1112133 0.5409 0.4633 Gender 1 1 0.0633231 0.3080 0.5798
Response Log HCC-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00026551 0.0049 0.9443
Age 1 1 0.08853004 1.6364 0.2030
Gender 1 1 0.03163332 0.5847 0.4458
Response Log HGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02700502 2.1107 0.1486
Age 1 1 0.00008849 0.0069 0.9338
Gender 1 1 0.03155367 2.4663 0.1186
Response Log HSP60 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.19081158 2.6162 0.1081
Age 1 1 0.00130405 0.0179 0.8938
Gender 1 1 0.27482413 3.7680 0.0543 Response Log 1-309 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00324637 0.0121
Age 1 1 0.61436763 2.2903
Gender 1 1 0.27417915 1.0221
Response Log ICAM-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06460391 3.3366
Age 1 1 0.06765591 3.4943
Gender 1 1 0.09470946 4.8915
Response Log IgA Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00560051 0.1144
Age 1 1 0.13078951 2.6709
Gender 1 1 0.00389298 0.0795
Response Log IgE Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.2880261 0.7577
Age 1 1 0.5265826 1.3852
Gender 1 1 3.7779222 9.9382
Response Log IGFBP-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.02609120 0.4563
Age 1 1 0.45355998 7.9321
Gender 1 1 0.00496559 0.0868
Response Log IgM Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.04135207 0.7445
Age 1 1 0.02000818 0.3602
Gender 1 1 0.02618176 0.4714
Response Log IL-10 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03805695 1.2132
Age 1 1 0.06472602 2.0633
Gender 1 1 0.00351746 0.1121
Response Log IL-12p40 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.10374307 2.4214 0.1220 Age 1 1 0.00768044 0.1793 0.6727 Gender 1 1 0.01550620 0.3619 0.5484
Response Log IL-13 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.48911995 10.7788
Age 1 1 0.04455125 0.9818
Gender 1 1 0.08630200 1.9018
Response Log IL-15 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06828949 1.6463
Age 1 1 0.06170346 1.4876
Gender 1 1 0.00943827 0.2275
Response Log IL-16 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00020927 0.0094
Age 1 1 0.01403645 0.6285
Gender 1 1 0.01030902 0.4616
Response Log IL-18 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.06469431 2.1718
Age 1 1 0.01594176 0.5352
Gender 1 1 0.03580745 1.2021 Response Log IL-1 ra Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.66953304 11.2399 0.0010 Age 1 1 0.00181344 0.0304 0.8617 Gender 1 1 0.05415893 0.9092 0.3420
Response Log IL-3 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.12863518 1.7065 0.1936 Age 1 1 0.27183199 3.6063 0.0597 Gender 1 1 0.07644101 1.0141 0.3157
Response Log IL-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.08454037 2.5767 0.1108 Age 1 1 0.02059001 0.6276 0.4296 Gender 1 1 0.03871825 1.1801 0.2793
Response Log IL-5 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.20577596 5.4678 0.0208 Age 1 1 0.01898051 0.5043 0.4788 Gender 1 1 0.05584317 1.4839 0.2253
Response Log IL-6 Receptor Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00029649 0.0168
Age 1 1 0.00328296 0.1856
Gender 1 1 0.00095843 0.0542
Response Log IL-7 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.80119996 8.9895 0.0032
Age 1 1 0.14740498 1.6539 0.2006
Gender 1 1 0.00790063 0.0886 0.7664
Response Log IL-8 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03424653 1.2286 0.2696 Age 1 1 0.16540503 5.9341 0.0161 Gender 1 1 0.08756335 3.1415 0.0786
Response Log Insulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.77207491 5.2419 0.0236 Age 1 1 0.32339507 2.1956 0.1407 Gender 1 1 0.76597402 5.2004 0.0241
Response Log IP-10 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.47718825 11.2782 0.0010 Age 1 1 0.99979975 23.6300 <.0001 Gender 1 1 0.20338323 4.8069 0.0300
Response Log Kidney Injury Molecule Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.24540713 2.0287 0.1566 Age 1 1 0.38962471 3.2209 0.0749 Gender 1 1 0.21381579 1.7676 0.1859
Response Log Leptin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.2102846 1.1542
Age 1 1 0.0559775 0.3072
Gender 1 1 5.6205406 30.8490
Response Log LH Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03608443 0.9000 0.3445 Age 1 1 0.02487082 0.6203 0.4323 Gender 1 1 0.70077750 17.4793 <.0001 Response Log Lipoprotein (a) Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.64010648 1.8548 0.1755 Age 1 1 0.01158812 0.0336 0.8549 Gender 1 1 0.33952815 0.9838 0.3230
Response Log MCP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.13273292 4.3513 0.0388
Age 1 1 0.00248396 0.0814 0.7758
Gender 1 1 0.01589563 0.5211 0.4716
Response Log MCP-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.56024410 14.1756 0.0002
Age 1 1 0.02406649 0.6089 0.4365
Gender 1 1 0.01171750 0.2965 0.5870
Response Log MCP-3 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.22430481 1.6098 0.2067
Age 1 1 0.01148317 0.0824 0.7745
Gender 1 1 0.15744123 1.1299 0.2897
Response Log MCP-4 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 1.3380887 13.1311 0.0004
Age 1 1 0.1033455 1.0142 0.3157
Gender 1 1 0.0069750 0.0684 0.7940
Response Log M-CSF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.01223382 1.6048 0.2074
Age 1 1 0.00164693 0.2160 0.6428
Gender 1 1 0.00317576 0.4166 0.5197
Response Log MDC Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00142175 0.0647 0.7996
Age 1 1 0.02180147 0.9917 0.3211
Gender 1 1 0.06813425 3.0994 0.0806 Response Log MIF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.05392112 0.7212 0.3973
Age 1 1 0.20658690 2.7630 0.0988
Gender 1 1 0.00458773 0.0614 0.8047
Response Log Gamma Interferon Induced Monokine Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.1217418 1.6536 0.2007 Age 1 1 2.7416123 37.2395 <.0001 Gender 1 1 0.4695848 6.3784 0.0127
Response Log MIP-1 alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04013824 2.2937 0.1322 Age 1 1 0.10221209 5.8408 0.0170 Gender 1 1 0.04973944 2.8423 0.0941
Response Log MIP-1 beta Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.35621626 13.3533 0.0004 Age 1 1 0.22454016 8.4172 0.0043 Gender 1 1 0.00009980 0.0037 0.9513
Response Log MIP-3 alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.07864725 1.5497 0.2153 Age 1 1 0.07718807 1.5210 0.2196 Gender 1 1 0.00208289 0.0410 0.8398 Response Log MMP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.37703553 10.1300 0.0018
Age 1 1 0.00114504 0.0308 0.8610
Gender 1 1 0.03285041 0.8826 0.3492
Response Log MMP-10 Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.04087544 2.1925 0.1410 Age 1 1 0.07204161 3.8642 0.0514 Gender 1 1 0.03161756 1.6959 0.1950
Response Log MMP-2 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00549654 0.4375 0.5095
Age 1 1 0.46914122 37.3392 <.0001
Gender 1 1 0.01350779 1.0751 0.3016
Response Log MMP-7 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00213612 0.0363 0.8492
Age 1 1 0.25108745 4.2659 0.0408
Gender 1 1 0.06678099 1.1346 0.2887
Response Log MMP9 (total) Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.02118275 0.4291 0.5135 Age 1 1 0.02350467 0.4762 0.4913 Gender 1 1 0.00014502 0.0029 0.9569
Response Log MMP-9 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.10829191 1.8615 0.1747 Age 1 1 0.00677530 0.1165 0.7334 Gender 1 1 0.00643394 0.1106 0.7400
Response Log Myeloid Progenitor Inhibitory Factor 1 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.10586132 2.3696 0.1260 Age 1 1 0.04753259 1.0640 0.3041 Gender 1 1 0.08751542 1.9590 0.1639 Response Log Myeloperoxidase Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.32541400 6.2139 0.0139 Age 1 1 0.00573308 0.1095 0.7413 Gender 1 1 0.04153126 0.7931 0.3747
Response Log Myoglobin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.0572154 1.0335 0.3112 Age 1 1 0.0748181 1.3514 0.2471 Gender 1 1 1.1977325 21.6340 <.0001
Response Log NGAL Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00005946 0.0026 0.9591 Age 1 1 0.00040492 0.0180 0.8935 Gender 1 1 0.04356045 1.9359 0.1664
Response Log NrCAM Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.05176007 1.7843 0.1839 Age 1 1 0.00168820 0.0582 0.8097 Gender 1 1 0.02007147 0.6919 0.4070
Response Log NT-proBNP Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0713455 0.5140 0.4746
Age 1 1 3.4520659 24.8705 <.0001
Gender 1 1 0.5734034 4.1311 0.0440 Response Log Osteopontin Effect Tests
Source N arm DF Sum of Squares F Ratio
DIAG 1 1 0.04777478 0.7799
Age 1 1 0.15210632 24829
Gender 1 1 0.00637867 0.1041
Response Log PAI-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 3.1379704 28.8223
Age 1 1 0.0378878 0.3480
Gender 1 1 0.0344729 0.3166
Response Log Pancreatic Polypeptide Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.16879415 1.3107 0.2543
Age 1 1 0.11647147 0.9044 0.3433
Gender 1 1 0.34910642 2.7107 0.1020
Response Log Prostatic Acid Phosphatase Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.0503131 1.3709 0.2437
Age 1 1 0.1821020 4.9617 0.0276
Gender 1 1 1.7033483 46.4106 <.0001
Response Log PAPP-A Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.03550076 1.1853 0.2782 Age 1 1 0.03452393 1.1527 0.2849 Gender 1 1 0.41685441 13.9179 0.0003
Response Log PARC Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00110612 0.0950 0.7584 Age 1 1 0.01737447 1.4921 0.2240 Gender 1 1 0.00000167 0.0001 0.9904
Response Log PDGF-BB Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 5.1181302 26.8029
Age 1 1 0.6215571 3.2550
Gender 1 1 0.0351862 0.1843
Response Log PLGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 1.5104813 18.1755 <.0001 Age 1 1 0.0047919 0.0577 0.8106 Gender 1 1 0.0152165 0.1831 0.6694
Response Log Progesterone Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.63537691 6.8566
Age 1 1 0.70500886 7.6080
Gender 1 1 0.79590559 8.5889
Response Log Proinsulin, Intact Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.30662306 3.2351
Age 1 1 0.63212507 6.6695
Gender 1 1 0.32759868 3.4564
Response Log Proinsulin, Total Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.35606379 3.4115
Age 1 1 0.52889525 5.0674
Gender 1 1 0.40346671 3.8656
Response Log Prolactin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02522257 0.9493 0.3316
Age 1 1 0.00001936 0.0007 0.9785
Gender 1 1 0.16903823 6.3623 0.0128 Response Clean Protein S Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.037866 0.0029
Age 1 1 0.480004 0.0363
Gender 1 1 22.592654 1.7082
Response Log PYY Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.20813797 3.2375 0.0742 Age 1 1 0.15276993 2.3763 0.1255 Gender 1 1 0.03604158 0.5606 0.4553
Response Log RANTES Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 5.1624809 26.9910 <.0001 Age 1 1 1.0057119 5.2582 0.0234 Gender 1 1 0.0699256 0.3656 0.5464
Response Log Resistin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03381765 1.2303
Age 1 1 0.04698379 1.7093
Gender 1 1 0.04391738 1.5977
Response Clean Serum Amyloid P Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 11.017618 0.4948 0.4830 Age 1 1 9.088078 04082 0.5240 Gender 1 1 41.314326 1.8555 0.1754
Response Log Stem Cell Factor Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.20542916 9.4332 0.0026
Age 1 1 0.00215016 0.0987 0.7538
Gender 1 1 0.00232134 0.1066 0.7446
Response Log SGOT Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00248217 0.2476 0.6196 Age 1 1 0.00000158 0.0002 0.9900 Gender 1 1 0.00524082 0.5228 0.4709
Response Log SHBG Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.12091206 2.8407 0.0942 Age 1 1 0.05331419 1.2526 0.2650 Gender 1 1 0.67269330 15.8043 0.0001
Response Log SOD Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.03006726 0.6490
Age 1 1 0.09787141 2.1126
Gender 1 1 0.00016725 0.0036
Response Log Sortilin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.09242051 9.9103 0.0020
Age 1 1 0.04792720 5.1393 0.0250
Gender 1 1 0.03575330 3.8339 0.0523
Response Log sRAGE Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02382189 0.4164 0.5198
Age 1 1 0.00079873 0.0140 0.9061
Gender 1 1 0.14626676 2.5567 0.1121
Response Log Tamm-Horsfall Protein Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.11212570 2.1025 0.1494
Age 1 1 0.16144443 3.0274 0.0841
Gender 1 1 0.13721515 2.5730 0.1110 Response Log Thyroxine Binding Globulin Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.00224600 0.3218 0.5715 Age 1 1 0.00085932 0.1231 0.7262 Gender 1 1 0.01889077 2.7066 0.1022
Response Log TECK Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.19896132 94176
Age 1 1 0.00247841 0.1173
Gender 1 1 0.00826751 0.3913
Response Log Tenasci Effect Tests
Source Nparm Sum of Squares F Ratio
DIAG 1 0.05174432 0.8593
Age 1 0.15720199 2.6106
Gender 1 0.00140192 0.0233
Response Log Testosterone Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.0186363 0.5271
Age 1 1 0.1762541 4.9855
Gender 1 1 7.0982949 200.7807
Response Log TGF-alpha Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 1.0100467 9.1677 0.0029
Age 1 1 0.0073251 0.0665 0.7969
Gender 1 1 0.0932355 0.8463 0.3592
Response Log Thrombomodulin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F DIAG 1 1 0.01356193 0.9942 0.3205 Age 1 1 0.00477802 0.3503 0.5549 Gender 1 1 0.05486106 4.0219 0.0469
Response Log TIMP-1 Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.08152942 6.3472
Age 1 1 0.09602086 7.4753
Gender 1 1 0.00179554 0.1398
Response Log TNFRII Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.00335240 0.1355
Age 1 1 0.31227349 12.6252
Gender 1 1 0.00108792 0.0440
Response Log Thrombopoietin Effect Tests
Source Nparm DF Sum of Squares F Ratio
DIAG 1 1 0.07698395 2.9021
Age 1 1 0.17209602 6.4875
Gender 1 1 0.02097030 0.7905
Response Log TRAIL R3 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.01820380 0.5790 0.4480
Age 1 1 0.00437378 0.1391 0.7097
Gender 1 1 0.08117603 2.5821 0.1104
Response Clean Transferrin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 288181.84 2.4253 0.1217
Age 1 1 325852.38 2.7424 0.1000
Gender 1 1 365861.43 3.0791 0.0816
Response Log Trefoil Factor 3 Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00023729 0.0073 0.9322
Age 1 1 0.32991606 10.1142 0.0018
Gender 1 1 0.10006665 3.0677 0.0821 Response Log TSH Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.10070560 1.3326 0.2504
Age 1 1 0.09927894 1.3138 0.2537
Gender 1 1 0.02936577 0.3886 0.5341
Response Log Thrombospondin-1 Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 5.6403855 25.3429 <.0001
Age 1 1 1.4831393 6.6639 0.0109
Gender 1 1 0.1794649 0.8064 0.3708
Response Clean TTR Effect Tests
Source N arm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 179.5341 0.0675 0.7954
Age 1 1 5044.5965 1.8963 0.1708
Gender 1 1 6300.4605 2.3684 0.1261
Response Log VCAM Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.00441269 0.5385 0.4643
Age 1 1 0.14738921 17.9860 <.0001
Gender 1 1 0.00541127 0.6603 0.4179
Response Log VEGF Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.02429722 0.8876 0.3478
Age 1 1 0.04232906 1.5463 0.2158
Gender 1 1 0.00289003 0.1056 0.7457
Response Clean Vitronectin Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 7.550 0.0007 0.9782
Age 1 1 16059.943 1.5905 0.2094
Gender 1 1 792.248 0.0785 0.7798
Response Log von Willebrand Factor Effect Tests
Source Nparm DF Sum of Squares F Ratio Prob > F
DIAG 1 1 0.33853498 5.6042 0.0193
Age 1 1 0.38269567 6.3352 0.0130
Gender 1 1 0.04314735 0.7143 0.3995
For purposes of the univariate analysis, analytes that had more than 10% missing values were excluded from the analysis. Values that were reported as LOW (below the lowest assay limit which is defined by RBM as a value below the lowest calibrator for the individual assay) were imputed by taking the reported low detection limit for individual assays (LDD) and dividing by 2. Individual analyte distributions were tested for normalcy using Box-Cox analysis. If necessary, analytes were log transformed and checked again for normalcy. The majority of analytes exhibited non-normal distributions and were log transformed. For purposes of the univariate analysis, multidimensional scaling and Mahalanobis distances were used to detect outliers. Outliers were defined as 5 standard deviations beyond the mean and were replaced with the 5 STD value. All Univariate results corrected for age and gender. A separate analysis also corrected for age, gender and ApoE. Table 10 shows Univariate results from UPenn Plasma Dataset. The results demonstrate that levels of some biomarkers were increased compared to a reference level. Whereas levels of other biomarkers were decreased compared to a reference level.
Table 10:
Analytes Analyte Analyte
Alpha 1 Antichymotrypsin† Fibrinogen† Pancreatic Polypeptide † 1
Alpha 1- Antitrypsin† G-CSF † PAI-I i
Alpha 2 Macroglobulin † GST i PDGF-BB i
Apolipoprotein H† GRO-alpha j Protein S†
Apolipoprotein D† 1-309† RANTES i
Apolipoprotein E J, IGF-BP2† Resistin†
Betacelluli n J, IL12p40 i Serum Amyloid P J,
CD40† IL-8† SGOT i
CD40 Ligand j IL-10 † SHBG†
CRP i IL-13† Sortilin J,
CKMB i IL-3† Tenascin C†
Cortisol† IgA† Thrombomodulin †
Cystatin C † MDC †
EGF i MIP1 beta j
EN-RAGE † MMP-7†
E-Selectin J, NGAL †
FAS† Osteopontin†
Table 1 1 shows Univariate results from UPenn CSF Dataset. The results demonstrate that levels of some biomarkers were increased compared to a reference level. Whereas levels of other biomarkers were decreased compared to a reference level.
Table 1 1 :
Figure imgf000093_0001
Fibrinogen J,
IGF-BP2†
Table 12 summarizes a performance model of AD versus Control with respect to the 24 top plasma analytes. Table 12 shows that some markers are increased and other markers are decreased as wells as how each marker is altered between plasma and CSF samples. It was also observed that certain markers correlate with other markers to provide an indication of disease state.
Table 12:
Figure imgf000094_0001
The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety.
While the invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

CLAIMS What is claimed is:
1. A composition for detecting Alzheimer's Disease in an individual with at least 80% sensitivity, comprising reagents for detecting a plurality of biomarkers for
Alzheimer's Disease in a biological sample from the individual.
2. The composition of claim 1 capable of detecting biomarkers of Alzheimer's disease in a plasma sample.
3. The composition of claim 1, wherein said biomarkers for Alzheimer's disease comprise Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
4. The composition of claim 1, wherein said biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE,
NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309.
5. The composition of claim 1, wherein said biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2 Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment), Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
6. The composition of claim 1, wherein said biomarkers is one or more biomarkers selected from Table 2.
7. The composition of claim 1, wherein the reagent is an antibody.
8. The composition of claim 1 or claim 7, wherein the reagent is on a solid support.
9. A method of diagnosing Alzheimer's Disease in an individual, the method comprising analyzing a biological sample from an individual to determine the level(s) of one or more biomarkers for Alzheimer's Disease in the sample, wherein the one or more biomarkers are selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to diagnose whether the individual has Alzheimer's Disease.
10. The method of claim 9, further comprising analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to diagnose whether the individual has Alzheimer's Disease.
11. The method of claim 10, wherein the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
12. The method of claim 1 1, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
13. The method of claim 9, wherein Alzheimer's Disease diagnosed is selected from the group consisting of late onset Alzheimer's disease, early onset
Alzheimer's disease, familial Alzheimer's disease, and sporadic Alzheimer's disease.
14. The method of claim 9, wherein said biological sample is selected from the group consisting of whole blood, a blood component, CSF, urine, and any combination thereof.
15. A method for determining whether dementia in an individual is associated with AD, the method comprising analyzing a biological sample from an individual to determine the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin in the sample, and comparing the level(s) of Cortisol, pancreatic polypeptide, osteopontin, IGF BP-2, and resistin in a corresponding reference sample to determine whether dementia in an individual is associated with AD.
16. The method of claim 15, wherein the dementia is associated with AD.
17. The method of claim 15 or 16, further comprising analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine the presence or status of dementia in the individual.
18. The method of claim 16, wherein the biological sample from the individual is analyzed to determine the level(s) of one or more biomarkers selected from Table 2; and comparing the level(s) of the one or more biomarkers in the sample to Alzheimer's Disease-positive and/or Alzheimer's Disease-negative reference levels of the one or more biomarkers in order to determine the presence or status of Alzheimer's Disease or other types of dementia in the individual.
19. The method of claim 18, wherein the levels of Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, resistin biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine the presence or status of
Alzheimer's Disease or other types of dementia in the individual.
20. The method of claim 19, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
21. A method for diagnosing Alzheimer's disease in an individual, the method comprising: measuring the level of one or more biomarkers selected from Table 2 in a plasma sample from the individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive control reference values and optionally to negative control reference values, wherein levels of the biomarkers in the plasma sample of the individual resemble the positive control reference values and do not resemble the negative control reference values are an indication that said individual has Alzheimer's disease.
22. The method of claim 21, further comprising analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the test individual in order to diagnose whether the individual has Alzheimer's Disease.
23. The method of claim 22, wherein the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm to diagnose whether the individual has Alzheimer's Disease.
24. The method of claim 23, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
25. A method of determining risk of an individual developing Alzheimer's disease, the method comprising:
measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual, and comparing the level of the biomarkers in the plasma sample of the individual to positive and optionally to negative control values, wherein levels of the biomarkers in the plasma sample of the individual that resemble the positive control and do not resemble the negative control are an indication that said individual is at risk of developing Alzheimer's disease.
26. The method of claim 25, further comprising analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to determine risk of an individual of developing Alzheimer's disease.
27. The method of claim 26, wherein the level of biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to determine risk of an individual of developing Alzheimer's disease.
28. The method of claim 27, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
29. A method for monitoring Alzheimer's disease in an individual, the method comprising:
measuring the level of one or more biomarkers selected from Table 2 in a plasma sample of an individual at a first time, and comparing the level of the biomarkers in the plasma sample of the individual at the first time to a level of the biomarkers in a plasma sample of the individual at the second time.
30. The method of claim 29, further comprising analyzing age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual in order to monitor Alzheimer's disease in the individual.
31. The method of claim 30, wherein the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to monitor Alzheimer's disease in the individual.
32. The method of claim 31, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
33. A method of assessing increased risk of developing Alzheimer's disease, said method comprising:
obtaining a first sample of plasma from an individual blood at a first time, assessing the level of a biomarker for Alzheimer's disease in said first plasma sample to obtain a baseline level,
obtaining a second sample of plasma from said individual at a second time to obtain a second level,
assessing the level of said biomarker in said second plasma sample to obtain a second level,
wherein if the second level is greater in the case of a biomarker that is over-expressed in AD or lower in the case of a biomarker that is under-expressed in AD, the individual is at an increased risk of developing Alzheimer's disease.
34. The method of claim 33, wherein the biomarker is one or more biomarkers selected from Table 2.
35. The method of claim 34, wherein age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual is analyzed in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
36. The method of claim 35 wherein the level of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess whether the individual is at an increased risk of developing Alzheimer's disease.
37. The method of claim 36, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares (PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
38. A method of assessing the likelihood that a pharmaceutical agent is efficacious in treating Alzheimer's disease in an individual, said method comprising:
obtaining a first sample of plasma from an individual in the absence of a pharmaceutical agent,
assessing the level of a biomarker for Alzheimer's disease in said first plasma sample to obtain a baseline level,
administering said pharmaceutical agent to said individual, obtaining a second sample of plasma from said individual after administration of said pharmaceutical agent,
assessing the level of said biomarker for Alzheimer's disease in said second plasma sample to obtain a treated level,
wherein the likelihood that the pharmaceutical agent treats Alzheimer's disease is increased if the treated level is lower than the baseline level in the case of a biomarker that is over-expressed in AD and higher than the baseline level in the case of a biomarker that is under-expressed in AD.
39. The method of claim 38, wherein the biomarker is one or more biomarkers selected from Table 2.
40. The method of claim 39, wherein age, gender, and ApoE genotype (e2/e2, e2/e3, e2/e4, e3/e3, e3/e4 and e4/e4) of the individual is analyzed in order to assess the likelihood that the pharmaceutical agent is efficacious in treating Alzheimer's disease in the individual.
41. The method of claim 40, wherein the levels of the biomarkers, age, gender, and ApoE genotype are incorporated into an algorithm in order to assess the likelihood that the pharmaceutical agent is efficacious in treating Alzheimer's disease in the individual.
42. The method of claim 41, wherein said algorithm is selected from the group consisting of Logistic Regression, Boosted Tree Models, Flexible Discriminant Analysis (FDA), K-Nearest Neighbors (KNN), Naive Bayes, Partial Least Squares
(PLS), Randon Forests, Shrunken Centroids, Sparse Partial Least Squares, Support Vector Machines approaches, and any combination thereof.
43. A method of diagnosing Alzheimer's Disease in an individual, comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and comparing said levels of each of said at least five biomarkers to non-AD controls, wherein a statistically significant increase in said levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in said level of ApoE provides a positive diagnosis of AD in said individual.
44. A method of diagnosing Alzheimer's Disease in an individual, comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and
comparing said levels of each of said at least five biomarkers to non-AD controls, wherein a statistically significant increase in said levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides a positive diagnosis of AD in said individual.
45. The method of claim 43 or 44, further comprising conducting one or more cognitive tests on said individual to confirm said positive diagnosis of AD.
46. The method of claim 43, 44 or 45, further comprising obtaining a CSF sample from said individual to confirm said positive diagnosis of AD.
47. A method for screening to identify individuals at increased risk of developing AD, the method comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and
comparing said levels of each of said at least five biomarkers to non-AD controls, wherein a statistically significant increase in said levels of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in said level of ApoE provides an identification of an individual at increased risk of developing AD.
48. A method for screening to identify individuals at increased risk of developing AD, the method comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and
comparing said levels of each of said at least five biomarkers to non-AD controls, wherein a statistically significant increase in said levels of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin provides an identification of an individual at increased risk of developing AD.
49. A method of diagnosing Alzheimer's Disease in an individual, comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and ApoE; and
determining whether, relative to non-AD controls, said levels of said at least five biomarkers provide a signature of a positive diagnosis of AD in said individual, wherein said signature comprises: a statistically significant increase in said levels of each of Cortisol, IGF BP2, pancreatic polypeptide, and resistin, and a statistically significant decrease in said level of ApoE.
50. A method of diagnosing Alzheimer's Disease in an individual, comprising
obtaining a plasma sample from said individual;
assaying levels of at least five biomarkers in said plasma sample, wherein said at least five biomarkers comprise: Cortisol, IGF BP2, pancreatic polypeptide, resistin, and osteopontin; and
determining whether, relative to non-AD controls, said levels of said at least five biomarkers provide a signature of a positive diagnosis of AD in said individual, wherein said signature comprises: a statistically significant increase in said levels of each of Cortisol, IGF BP2, pancreatic polypeptide, osteopontin and resistin.
51. A kit for detecting Alzheimer's Disease in an individual with at least 80% sensitivity, comprising a reagent for detecting a plurality of biomarkers for Alzheimer's Disease in a biological sample from the individual.
52. The kit of claim 51 , wherein said reagent is capable of detecting biomarkers of Alzheimer's disease in a plasma sample.
53. The kit of claim 51, wherein said biomarkers of Alzheimer's disease comprises Cortisol, pancreatic polypeptide, osteopontin, IGF BP2, and resistin.
54. The kit of claim 51, wherein said biomarkers for Alzheimer's disease comprise resistin, pancreatic polypeptide, ApoD, G-CSF, MlPlbeta, ApoE, NrCAM, MMP2, Cortisol, PDGF-BB, MMP1, and 1-309.
55. The kit of claim 51, wherein said biomarkers for Alzheimer's disease comprise Alpha- 1 Microglobulin, Angiopoietin-2, Apolipoprotein E, Beta-2
Microglobulin, Chemokine (C-X-C motif) ligand 13 (CXCL13) also known as B lymphocyte chemoattractant (BLC), Cortisol, E-Selectin, FAS, Fatty Acid Binding Protein, Hepatocyte Growth Factor, IGF BP-2, Interleukin 10, NT-Pro-BNP (Brain natriuretic peptide co-secreted along with a 76 amino acid N-terminal fragment),
Osteopontin, Pancreatic Polypeptide, PAPP-A, Resistin, Stem Cell Factor, Tenascin C, Thrombomodulin, tissue inhibitor of metalloproteinases-1 (TIMP-1), vascular cell adhesion molecule- 1 (VCAM-1), Vascular endothelial growth factor (VEGF), and Von Willebrand Factor.
56. The kit of claim 51, wherein said biomarkers is one or more biomarkers selected from Table 2.
57. The kit of claim 51, wherein the reagent is an antibody.
58. The kit of claim 51 or claim 57, wherein the reagent is on a solid support.
The kit of claim 51 or 57 comprising an instruction manual.
PCT/US2011/036457 2010-05-14 2011-05-13 Plasma biomarkers for diagnosis of alzheimer's disease WO2011143574A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33489810P 2010-05-14 2010-05-14
US61/334,898 2010-05-14

Publications (2)

Publication Number Publication Date
WO2011143574A2 true WO2011143574A2 (en) 2011-11-17
WO2011143574A3 WO2011143574A3 (en) 2012-05-18

Family

ID=44359502

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/036457 WO2011143574A2 (en) 2010-05-14 2011-05-13 Plasma biomarkers for diagnosis of alzheimer's disease

Country Status (1)

Country Link
WO (1) WO2011143574A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140228240A1 (en) * 2013-02-14 2014-08-14 Emory University Screening Blood for Protein Biomarkers and Uses Thereof in Alzheimer's Disease and Mild Cognitive Impairment
WO2015193427A1 (en) * 2014-06-19 2015-12-23 Olink Ab Determination and analysis of biomarkers in clinical samples
WO2016081100A1 (en) * 2014-11-20 2016-05-26 Ge Healthcare Uk Limited Detecting dementia and alzheimer's disease associated biomarkers stabilized on solid support materials
EP3067699A1 (en) 2015-03-11 2016-09-14 Neuron Bio, S.A. Method for diagnosing alzheimer's disease
EP3376230A1 (en) 2017-03-13 2018-09-19 Biocross, S.L. Identification of signatures for neurodegeneration diseases diagnoses
RU2687272C2 (en) * 2014-01-28 2019-05-13 Предемтек Аг Biomarker and methods for early diagnosis of alzheimer's disease
WO2019215085A1 (en) 2018-05-07 2019-11-14 Biotx.Ai Gmbh Method for predicting the risk of late-onset alzheimer's diseases
US10670611B2 (en) 2014-09-26 2020-06-02 Somalogic, Inc. Cardiovascular risk event prediction and uses thereof
WO2022120373A1 (en) * 2020-12-03 2022-06-09 Amgen Inc. Trem2 agonist biomarkers and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5286262A (en) 1987-03-02 1994-02-15 Atrium Medical Corp. Multipurpose collection vessel
US5654152A (en) 1988-02-19 1997-08-05 Showa Denko Kabushiki Kaisha Method of measuring enzyme activity by using a column having an immobilized substrate
US20030224490A1 (en) 2001-12-18 2003-12-04 Whitehead Institute For Biomedical Research Fusion partner cells and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5286262A (en) 1987-03-02 1994-02-15 Atrium Medical Corp. Multipurpose collection vessel
US5654152A (en) 1988-02-19 1997-08-05 Showa Denko Kabushiki Kaisha Method of measuring enzyme activity by using a column having an immobilized substrate
US20030224490A1 (en) 2001-12-18 2003-12-04 Whitehead Institute For Biomedical Research Fusion partner cells and uses thereof

Non-Patent Citations (50)

* Cited by examiner, † Cited by third party
Title
"Molecular and Biochemical Markers of Alzheimer's Disease", THE RONALD AND NANCY REAGAN RESEARCH INSTITUTE OF THE ALZHEIMER'S ASSOCIATION AND THE NATIONAL INSTITUTE ON AGING WORKING GROUP. NEUROBIOL AGING., vol. 19, no. 2, March 1998 (1998-03-01), pages 109 - 116
ANNALS OF STATISTICS, 2001, pages 1189 - 1232
BARBAS, NATURE MEDICINE, vol. 1, 1995, pages 837 - 839
BENSON ET AL., NUCLEIC ACIDS RESEARCH, 2008
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BREIMAN: "Random forests", MACHINE LEARNING, vol. 45, no. 1, 2001, pages 5 - 32
BURTON ET AL., ADV. IMMUNOL., vol. 57, 1994, pages 191 - 280
CHUN H, KELES S: "Sparse partial least squares for simultaneous dimension reduction and variable selection", JOURNAL OF THE ROYAL STATISTICAL SOCIETY, vol. 182, no. 1, 2009, pages 79 - 90
CRAIG-SCHAPIRO ET AL., NEUROBIOL DIS, vol. 35, no. 2, 2009, pages 128 - 140
DE KRUIF ET AL., J. MOL. BIOL., vol. 248, 1995, pages 97 - 105
DUBOIS ET AL., LANCET NEUROL., vol. 6, no. 8, 2007, pages 734 - 746
FARRER ET AL., JAMA, vol. 278, no. 16, 1997, pages 1349 - 1356
FOLSTEIN ET AL., J. PSYCHIATR. RES, vol. 12, 1975, pages 1289 - 198
GATZ ET AL., ARCH. GEN. PSYCHIATRY, vol. 63, no. 2, 2006, pages 168 - 174
GU ET AL., THROMBOSIS AND HEMATOCYST, vol. 77, no. 4, 1997, pages 755 - 759
HARLOW ET AL.: "Antibodies, A Laboratory Manual", 1988, COLD SPRING HARBOR
HARLOW ET AL.: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR
HARLOW ET AL.: "Antibodies: A Laboratory Manual", 1989, COLD SPRING HARBOR
HARLOW ET AL.: "Using Antibodies: A Laboratory Manual", 1999, COLD SPRING HARBOR LABORATORY PRESS
HASTIE ET AL.: "Flexible Discriminant Analysis by Optimal Scoring", JOURNAL OF THE AMERICAN STATISTICAL ASSOCIATION, vol. 89, 1994
HIRSCH-REINSHAGEN ET AL., MOL CELL BIOCHEM., vol. 326, no. 1-2, 2009, pages 121 - 129
HOUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
HYE ET AL., BRAIN, vol. 129, 2006, pages 3042 - 3050
IRIZARRY ET AL., NEURORX, vol. 1, no. 2, 2004, pages 226 - 234
JACK ET AL., BRAIN, vol. 132, 2009, pages 1355 - 1365
KIM ET AL., NEURON, vol. 63, no. 3, 2009, pages 287 - 303
LANDAU ET AL., NEUROBIOL AGING., 4 April 2009 (2009-04-04)
LEOW ET AL., NEUROIMAGE, 2009
M. ILYAS KAMBOH: "Molecular Genetics of Late-Onset Alzheimer's Disease", ANNALS OF HUMAN GENETICS, vol. 68, no. 4, 2004, pages 381 - 404
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MARTENS, NAESES: "Multivariate calibration.", 1989, WILEY
MATTSSON ET AL., JAMA, vol. 302, no. 4, 2009, pages 385 - 393
MCKHANN ET AL., NEUROLOGY, vol. 34, no. 7, 1984, pages 939 - 944
PETERSEN ET AL., CNS SPECTR., vol. 13, no. 1, 2008, pages 45 - 53
RAY ET AL., NAT MED., vol. 13, no. 11, 2007, pages 1359 - 1362
RAY ET AL., NAT MERL., vol. 13, no. 11, 2007, pages 1359 - 1362
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
SAUNDERS ET AL., NEUROLOGY, vol. 43, no. 8, 1993, pages 1467 - 1472
SAVVA ET AL., N ENGL J MED., vol. 360, no. 22, 2009, pages 2302 - 2309
SHAW ET AL., ANN NEUROL., vol. 65, no. 4, 2009, pages 403 - 413
SONG ET AL., BRAIN RES REV., 2009
SUNDERLAND ET AL., JAMA, vol. 289, no. 16, 2003, pages 2094 - 2103
TIBSHIRANI ET AL.: "Class prediction by nearest shrunken centroids, with applications to DNA microarrays", STATISTICAL SCIENCE, 2003, pages 104 - 117
TREVOR HASTIE, ROBERT TIBSHIRANI, JEROME FRIEDMAN: "The Elements of Statistical Learning: Data Mining, Inference, and Prediction", 2009
TUSHER ET AL., PROC NATL ACAD SCI U S A., vol. 98, no. 9, 2001, pages 5116 - 5121
TUSZYNSKI ET AL., BLOOD, vol. 72, 1988, pages 109 - 115
VAPNIK ET AL., IEEE TRANS NEURAL NETW., vol. 10, no. 5, 1999, pages 988 - 999
VISSER ET AL., LANCET NEUROL., vol. 8, no. 7, 2009, pages 619 - 627
WRIGHT ET AL., CRITICAL REV. IMMUNOL., vol. 12, no. 3, 4, 1992, pages 125 - 168
ZHANG ET AL., PROTEOMICS, vol. 4, no. 1, pages 244 - 256

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140228240A1 (en) * 2013-02-14 2014-08-14 Emory University Screening Blood for Protein Biomarkers and Uses Thereof in Alzheimer's Disease and Mild Cognitive Impairment
RU2687272C2 (en) * 2014-01-28 2019-05-13 Предемтек Аг Biomarker and methods for early diagnosis of alzheimer's disease
US10352949B2 (en) 2014-01-28 2019-07-16 Predemtec Ag Biomarker and methods for early diagnosis of Alzheimer's disease
WO2015193427A1 (en) * 2014-06-19 2015-12-23 Olink Ab Determination and analysis of biomarkers in clinical samples
US10670611B2 (en) 2014-09-26 2020-06-02 Somalogic, Inc. Cardiovascular risk event prediction and uses thereof
WO2016081100A1 (en) * 2014-11-20 2016-05-26 Ge Healthcare Uk Limited Detecting dementia and alzheimer's disease associated biomarkers stabilized on solid support materials
CN107076764A (en) * 2014-11-20 2017-08-18 通用电气医疗集团英国有限公司 The stable dementia and Alzheimer disease relevant biomarkers on solid carrier material of detection
JP2018502281A (en) * 2014-11-20 2018-01-25 ジーイー・ヘルスケア・ユーケイ・リミテッド Detection of dementia and Alzheimer's disease related biomarkers immobilized on solid support material
US20180031575A1 (en) * 2014-11-20 2018-02-01 Ge Healthcare Uk Limited Detecting Dementia and Alzheimer's Disease Associated Biomarkers Stabilized on Solid Support Materials
EP3067699A1 (en) 2015-03-11 2016-09-14 Neuron Bio, S.A. Method for diagnosing alzheimer's disease
WO2016142450A1 (en) 2015-03-11 2016-09-15 Neuron Bio, S.A. Method for diagnosing alzheimer's disease
WO2018166893A1 (en) 2017-03-13 2018-09-20 Biocross, S.L. Identification of signatures for neurodegeneration diseases diagnoses
EP3376230A1 (en) 2017-03-13 2018-09-19 Biocross, S.L. Identification of signatures for neurodegeneration diseases diagnoses
WO2019215085A1 (en) 2018-05-07 2019-11-14 Biotx.Ai Gmbh Method for predicting the risk of late-onset alzheimer's diseases
WO2022120373A1 (en) * 2020-12-03 2022-06-09 Amgen Inc. Trem2 agonist biomarkers and methods of use thereof

Also Published As

Publication number Publication date
WO2011143574A3 (en) 2012-05-18

Similar Documents

Publication Publication Date Title
AU2018201062B2 (en) Personalized medicine approach for treating cognitive loss
WO2011143574A2 (en) Plasma biomarkers for diagnosis of alzheimer&#39;s disease
Hansson et al. Blood biomarkers for Alzheimer’s disease in clinical practice and trials
EP2965090B1 (en) Methods and compositions for the diagnosis of alzheimer&#39;s disease
US20180106817A1 (en) Protein biomarkers and therapeutic targets for renal disorders
AU2016266053A1 (en) Method for detection of a neurological disease
JP2018513368A (en) A method for predicting the risk of cognitive decline
US20130116135A1 (en) Methods, Kits and Reagents for Diagnosing, Alding Diagnosis and/or Monitoring Progression of a Neurological Disorder
EP3608424A2 (en) Blood biomarker for discerning beta amyloid accumulation in brain
US20140228240A1 (en) Screening Blood for Protein Biomarkers and Uses Thereof in Alzheimer&#39;s Disease and Mild Cognitive Impairment
US10739355B2 (en) Serum biomarker panels for bipolar disorder
US20190277864A1 (en) Methods for detecting an individual as being at risk of developing a neurodegenerative disease
WO2021009074A1 (en) Novel markers as early predictors of alzheimer&#39;s pathology
Li et al. Cerebrospinal fluid α-synuclein predicts neurodegeneration and clinical progression in non-demented elders
KR102254053B1 (en) Biomarker for detecting amyloid beta accumulation in brain of subject with normal cognitive function or mild cognitive impairment using blood sample
CN115461474A (en) Protein markers for assessing alzheimer&#39;s disease
Liu et al. Periphery biomarkers for objective diagnosis of cognitive decline in type 2 diabetes patients
EP2690440A1 (en) Protein level of C9ORF72 for diagnosing a neurodegenerative disease
Gaetani et al. Required improvements for cerebrospinal fluid-based biomarker tests of Alzheimer’s disease
EP4332575A1 (en) Methods for diagnosis and prognosis of alzheimer&#39;s disease
US20240003918A1 (en) Non-invasive assessment of alzheimer&#39;s disease
WO2011109503A1 (en) Novel csf biomarkers for alzheimer&#39;s disease and frontotemporal lobar degeneration
Sakkaki et al. Gene expression patterns of CRYM and SIGLEC10 in Alzheimer's disease: potential early diagnostic indicators
Jijing et al. Deamidation-related blood biomarkers show promise for early diagnostics of neurodegeneration
KR20230081333A (en) Method for providing information concerning diagnosis of alzheimer&#39;s diseases

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11724089

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11724089

Country of ref document: EP

Kind code of ref document: A2