Methods and Compositions for the Diagnosis and Treatment of Kawasaki Disease

Abstract
The present invention relates to the determination of levels or expression of particular biomarkers in biological samples which can be utilized to diagnose, prognose, and treat Kawasaki disease in subjects, and further to select subjects who would benefit from a Kawasaki disease therapy other than, or in addition to, IVIG treatment. Accordingly, the present invention encompasses methods and compositions that utilize these biomarkers for the diagnosis, prognosis, and treatment of Kawasaki disease.
Description
BACKGROUND

Kawasaki disease is an acute, systemic vasculitis predominantly affecting young children. Clinical symptoms of Kawasaki disease include persistent fever not managed by antipyretic medications and antibiotics, rash, conjunctival infection, edema and erythema of the extremities, and oropharyngeal erythema.


The disease may impact the systemic vasculature, but the coronary arteries and cardiac tissue are particularly susceptible to damage. Inflammation of the coronary arteries and surrounding cardiac tissue may be mild and reversible or may be extensive, leading to cardiac artery aneurysms (ballooning) and stenosis (narrowing) of the arteries. An estimated 25% of Kawasaki disease subjects develop cardiac artery aneurysms or stenosis. While many subjects show recovery of cardiac functions and no angiographic evidence of cardiac artery aneurysms or stenosis following recovery, there can be evidence of continued endothelial and vascular dysfunction even years later. Subjects with larger aneurysms are at higher risk for myocardial infarct (MI) and other cardiovascular events later in life.


There is no specific test available to diagnose Kawasaki disease. Diagnosis largely is a process of ruling out diseases that cause similar signs and symptoms (http://www.mayoclinic.org/diseases-conditions/kawasaki-disease/basics/tests-diagnosis/con-20024663, Mar. 3, 2014).


SUMMARY OF THE INVENTION

Applicants have discovered that determination of levels or expression of particular biomarkers (e.g., protein levels, mRNA levels, glycan abundance, and/or the binding properties of IgG) in biological samples can be utilized to diagnose, prognose, and treat Kawasaki disease in subjects, and further to select subjects who would benefit from a Kawasaki disease therapy other than, or in addition to, IVIG treatment. Accordingly, the present invention encompasses methods and compositions that utilize these proteins for the diagnosis, prognosis, and treatment of Kawasaki disease.


In a first aspect, the invention features a method for diagnosing Kawasaki disease in a subject. This method includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) proteins of Table 1 and/or Table 2 e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more and/or determining whether IgG in the sample binds to one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, forty, fifty, sixty, or more) peptides of Table 3.









TABLE 1





Selected Protein


Biomarkers


Protein Name







Alpha-1-antichymotrypsin


(serpin peptidase inhibitor,


clade A, member 3)


lactate dehydrogenase C


armadillo repeat


containing 2


unc-45 homolog A


(C. elegans)


defensin,


alpha 1


S100 calcium


binding


protein A9


solute carrier family 26


(anion exchanger),


member 3


zeta-chain (TCR)


associated protein


kinase 70 kDa


Zinc finger protein


106 homolog


apolipoprotein B


Zinc finger protein


161 homolog


apolipoprotein M


apolipoprotein C-II


apolipoprotein F


zinc finger protein 578


xin actin-binding


repeat containing 2


lipoprotein, Lp(a)


armadillo repeat


containing 10


olfactory receptor,


family 5, subfamily D,


member 14


orosomucoid 1


Alpha-1-antitrypsin


(serpin peptidase inhibitor,


clade A, member 1)


nitric oxide synthase 2,


inducible (Gene)


carcinoembryonic


antigen-related cell


adhesion molecule 8


(Gene)


interleukin 33 (Gene)


interleukin 6 (interferon,


beta 2) (Gene)


matrix metallopeptidase 1


(interstitial collagenase)


(Gene)
















TABLE 2





Selected Protein


Biomarkers


Protein Name







collagen, type


VI, alpha 3


F-box protein 47


collagen, type


VI, alpha 1


androgen receptor


tumor protein p53


binding protein 1


peptidase inhibitor 16


cadherin 13


tenascin XB


peroxisome


proliferator-activated


receptor alpha


collagen, type I, alpha 1


Ig alpha-2 chain C


region (A2m marker)


Ig heavy chain


V-III region TIL


Ig alpha-1


chain C region


olfactomedin 1


complement


factor H


prostaglandin D2


synthase 21 kDa


(brain)


sex hormone-


binding globulin


Ig kappa


chain V-I region


gelsolin


Ig lambda-1 chain C


regions (Mcg marker)


BMP2 inducible kinase


antithrombin III


(serpin peptidase


inhibitor, clade C,


member 1)


pregnancy-zone protein


inter-alpha-trypsin


inhibitor heavy chain 1


Ig kappa chain V-II region


inter-alpha-trypsin


inhibitor heavy chain 2


complement factor H


Hepatocyte growth


factor activator


attractin


kininogen 1


coagulation factor


XIII, B polypeptide


complement


factor B


complement


component 1,


r subcomponent


extracellular matrix


protein 1


complement


component 1,


s subcomponent


complement


component 6


vitamin D binding


protein (group-


specific component)
















TABLE 3







Selected IgG Binding Peptides










SEQ

SEQ



ID NO.
Peptide Sequence
ID NO.
Peptide Sequence





 1
AKFLGQSTYIAGYHQVD
35
AMLGGMWAAYYPFPVPG





 2
FWSKMKPSEEYTTFYRD
36
YYWATGPEGPFRHPGAR





 3
FDRSDYMSFHLDDNITI
37
DMEFTVFDIDMEKHYKY





 4
IRIETPYYKDTEDGKYF
38
HYWDYQQLGFQGHLDHR





 5
LGLLQAITRNSWVDSAF
39
DQHFVWGPTGRAPMNYG





 6
KHWEFMQFDIGYIYEKF
40
TNGFHIPFYSDFQSAAA





 7
NIPSNQHATEIQVDGYH
41
MKYDVWKFYNGDDMRVS





 8
ENFEYHLYDSMIGYEVH
42
WYDNMYKTGFYRMYLLT





 9
ERPDPATYFMPGRDDQY
43
EFQDYNHSDVNMSNHPY





10
DNAPYYYREEWHKEFNK
44
YIPEQQPHEADNLYKDA





11
YHWDVQNTFYSMLMLPS
45
FYTLPHRPLYYYGYVAS





12
GDFSDYAPTLTQKASYG
46
HRWLEEANTEYMTMNSI





13
QYAFHNLDONGTVFGNR
47
GPINAYQKQDYSIEPEH





14
YVTNMMINMNYSSLSYS
48
AGDQYVRIDKSTRISNI





15
HWVLSDGYREVYSYNSY
49
SMKTVEADWYTYEPWWH





16
AYHSQLYIDYKDTEWFY
50
NHIYIVQTAYGVTGETS





17
DHPYFVIWDRYKPVHTY
51
MFAYHRAWPVWSSVLHV





18
HEHPPYLGMTAYELAQD
52
WPNPYFYHKKDTYWAHY





19
GQWSGQGYWYDPFDNMK
53
NYDHLLGQYPIRNWWSL





20
TTHFLKDRFESTNHDVY
54
ESMDVVWPYGYKFTQYW





21
VEDPRVGHSLFQDANYY
55
THMEHDFHIPLEMYKYM





22
DPVQIFNTAEHSGPYIR
56
VTPLEIIEQIREHLDIK





23
HDHFRGGKFILSTQAIW
57
LQKPFDYYMKEWQVDNE





24
NYPLPKYYYNWFEPRVW
58
VMQRWPENHFLQTHYDD





25
QYDDPDWQIHYKLEARG
59
WWNWRSAYKEGDVAYPS





26
QDPYMDLHYDKNQIEQA
60
GWVLDDWSSHHINYYIE





27
RGPHNFEIAETDAQMIE
61
FSPHYQYVGLFPYVKYI





28
DAYTNQDISEEEHMHRY
62
ANLVYMWGSAVHTSDPQ





29
GALEWIYYAGPKPGYWE
63
SPVMYSIANYKYQTMHL





30
PHSTHQIFYKSYETDMA
64
DQEISYLNSHTDLFVGR





31
IYWGPMSTGHLPSQAQF
65
YEPTDVYLTYRKLATKD





32
YTHFWWLDKYMRYEVAT
66
PVPQERTDLFTGAHRAL





33
FLTEYYEYQNNLFHAFR
67
REVYHEIKSGRAIEIYM





34
SRGDAAAWGILFDANWK
68
HDAIWYDWNPYPSKHES









According to this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of at least one protein of Table 1, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of at least one protein of Table 2, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of IgG in said sample to a peptide of Table 3, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative of the subject having Kawasaki disease.


In some embodiments, the method further includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) additional biomarkers in the biological sample. In certain embodiments, the one or more additional biomarkers are a protein of Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9; an mRNA of Table 10, Table 11, Table 12, and/or Table 13; and/or a glycan of Table 14, Table 15, Table 16, and/or Table 17.









TABLE 4





Selected Protein Biomarkers







L-lactate dehydrogenase


CD44 antigen


Ankyrin repeat domain-


containing protein 26


Lysosome-associated


membrane glycoprotein 2


Basal cell adhesion molecule


Multimerin-1


Transforming growth factor-


beta-induced protein ig-h3


Methylcytosine dioxygenase TET2


Alpha-1-antichynnotrypsin


Cystatin-C


Aftiphilin


Death domain containing 1


Xin actin-binding repeat containing 2


Metastasis associated 1


family, member 3
















TABLE 5





Selected Protein Biomarkers







Plasminogen


Vitamin K-dependent protein C


Coagulation factor XI


Apolipoprotein F


HGF activator


Bone marrow stromal


cell antigen 1


Mannan-binding lectin


serine peptidase 2


HCG2014417, isoform CRA a


T-lymphoma invasion and


metastasis-inducing protein 1
















TABLE 6





Selected Protein Biomarkers







IgA


Hemoglobin subunit gamma-1


Hemoglobin subunit gamma-2


Protein S100-A9


Protein S100-A8


Leucine-rich alpha-


2-glycoprotein


orosomucoid 1


orosomucoid 2


Actin, cytoplasmic 1


Haptoglobin-related protein


Cathepsin D


C-reactive protein


Xin actin-binding


repeat containing 2


Beta-galactoside alpha-


2,6-sialyltransferase 1


Complement component 9


Haptoglobin


Matrix metallopeptidase 9


Nidogen-1
















TABLE 7





Selected Protein Biomarkers







Gelsolin


Kallistatin


Kininogen-1


Lumican


Alpha-2-HS-glycoprotein


butyrylcholinesterase


Coagulation factor IX


Protein C


Ig heavy chain


V-III region BRO


Selenoprotein P


Cadherin-13


Myosin-8


Androgen receptor


Afamin


Biotinidase


Neural cell adhesion


molecule L1-like protein


Coagulation factor X


Tenascin XB


Titin
















TABLE 8





Selected Protein Biomarkers







Olfactory receptor, family 5,


subfamily D, member 14


Orosomucoid 2


Polycystin (PKD) family


receptor for egg jelly


Serpin peptidase inhibitor,


clade A (alpha-1 antiproteinase,


antitrypsin), member 3


Serpin peptidase inhibitor,


clade A (alpha-1 antiproteinase,


antitrypsin), member 1


Complement component 9


C-reactive protein,


pentraxin related


Haptoglobin-related protein


Haptoglobin


Orosomucoid 1


Leucine-rich alpha-2-glycoprotein


Protein S100-A9


Lipopolysaccharide binding protein


Beta-galactoside alpha-


2,6-sialyltransferase 1


Hemaglobin, alpha 1


Protein S100-A8


Hemoglobin, delta


Fc fragment of IgG, low


affinity IIIa receptor (CD16a)


Hemaglobin, gamma G


Hemaglobin, gamma A


Hemaglobin, beta


Kinesin family member 20B


Lysozyme


Actin, alpha, cardiac muscle 1


Inter-alpha-trypsin inhibitor


heavy chain 3


Inter-alpha-trypsin inhibitor


heavy chain family, member 4


Actin, beta-like 2


Hemaglobin, epsilon 1


IgA


Protein phosphatase 6,


regulatory subunit 2


Dedicator of cytokinesis 3


Cathepsin D


Centrosomal protein 290 kDa
















TABLE 9





Selected Protein Biomarkers







Complement component 2


Complement factor H-related 3


Clusterin


Kallikrein B, plasma (Fletcher factor) 1


Macrophage stimulating 1


(hepatocyte growth factor-like)


Biotinidase


Peptidoglycan recognition protein 2


Fibulin 1


Kininogen 1


Gelsolin


Mannan-binding lectin


serine peptidase 1


(C4/C2 activating component


of Ra-reactive factor)


Alpha-1-microglobulin/bikunin precursor


Apolipoprotein H (beta-2-glycoprotein I)


Sex hormone-binding globulin


Collagen, type I, alpha 1


Inter-alpha-trypsin inhibitor heavy chain 2


Inter-alpha-trypsin inhibitor heavy chain 1


TBC1 domain family, member


8B (with GRAM domain)


Afamin


Heat shock 70 kDa protein 5


(glucose-regulated protein, 78 kDa)


BMP2 inducible kinase


Collagen, type VI, alpha 3


Kallistatin


Lumican


Cadherin 13


Neural cell adhesion


molecule L1-like protein


Androgen receptor


Hepatocyte growth factor activator


Insulin-like growth factor


binding protein, acid labile subunit


Tenascin XB


Alpha-2-HS-glycoprotein


Glycosylphosphatidylinositol


specific phospholipase D1


Retinol binding protein 4, plasma


Serpin peptidase inhibitor,


clade C (antithrombin), member 1


butyrylcholinesterase


Complement factor H


Centrosomal protein 70 kDa


Coagulation factor XIII,


B polypeptide


Thrombospondin 4


Fibronectin 1


Insulin-like growth


factor 2 (somatomedin A)


Protein C


Selenoprotein P, plasma 1


Neuropilin 1


Neural cell adhesion molecule 1


alpha-1-B glycoprotein


Coagulation factor XIII,


A1 polypeptide


Extracellular matrix protein 1


Apolipoprotein C-1


Carboxypeptidase B2 (plasma)


Attractin


Zinc finger protein 217


Sex hormone-binding globulin


Pregnancy zone protein


Collagen, type XI, alpha 2


Selectin L


Ectonucleotide pyrophosphatase/


phosphodiesterase 2


Coagulation factor 11 (thrombin)


Apolipoprotein D


Plasmoginen


Insulin growth factor binding protein 3


Dehydrogenase/reductase


(SDR family) member 11


Quiescin Q6 sulfhydryl oxidase 1
















TABLE 10





Selected mRNA Biomarkers







CD80


Beta-glucuronidase
















TABLE 11





Selected mRNA Biomarkers







carcinoembryonic antigen-related


cell adhesion molecule 8


S100 calcium binding protein A12


Versican
















TABLE 12





Selected mRNA Biomarkers







Fc fragment of IgG, high affinity Ia, receptor (CD64)


S100 calcium binding protein A12


Matrix metallopeptidase 9


Tumor necrosis factor receptor superfamily, member 1A


Versican


Colony stimulating factor 2 receptor, beta, low affinity


(granulocyte-macrophage)


Interleukin 1, beta


Interleukin 1 receptor antagonist


S100 calcium binding protein A8


S100 calcium binding protein A11


Tumor necrosis factor receptor superfamily, member 1B


Tumor necrosis factor (ligand) superfamily, member 13b


Fc fragment of IgG, low affinity lia receptor (CD32)


Chemokine (C-C motif) receptor 2


Actin, beta


Tumor necrosis factor
















TABLE 13





Selected mRNA Biomarkers







Beta-glucuronidase


CD80


Killer cell lectin-like receptor subfamily G, member 1
















TABLE 14





Selected IgG and IgG Glycan Biomarkers







GP38HA


GP23HA


GP41HA


GP28HA


GP37HA


GP42HA


GP43HA


GP43LA


GP44HA


GP44LA


GP9HA


GP9LA
















TABLE 15





Selected IgG and IgG Glycan Biomarkers







GP6HA


GP4HA


GP26HA


GP11HA


GP5HA


GP7HA


GP2HA


GP12HA


IgG2_G0F_BGIcNAc


IgG2_BGIcNAc


GP3HA


IgG2_G1F_BGIcNAc


GP13HA


GP10HA


IgG3_4_A1


GP12LA


GP14HA


GP14LA


GP16LA


GP18HA


GP18LA


GP19LA


GP20HA


GP20LA


GP24LA


GP29LA


GP32HA


GP32LA


GP34HA


GP34LA


GP38LA


GP3LA
















TABLE 16





Selected IgG and IgG Glycan Biomarkers







GP41LA


GP42LA


GP42HA


GP41HA


GP43LA


GP43HA


GP37HA


GP44HA


GP44LA


GP23HA


GP35HA


GP31HA


IgA_G1_S1
















TABLE 17





Selected IgG and IgG Glycan Biomarkers







GP13HA


GP18HA


GP12LA


GP20LA


GP18LA


GP12HA


GP11HA


GP19HA


GP3HA


GP10HA


GP5HA


GP6HA


GP13LA


GP24LA


GP8HA


GP7HA


GP32LA


GP26HA


GP5LA


GP19LA


GP6LA


GP2HA


GP20HA









By this method, a subject can be further diagnosed for a predisposition to develop a secondary Kawasaki disease symptom, e.g., a cardiac artery aneurysm or stenosis. According to this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) is indicative of said subject having a predisposition to develop a secondary Kawasaki disease symptom, e.g., a cardiac artery aneurysm or stenosis.


In a second aspect, the invention features a method for diagnosing whether a subject has a predisposition to develop cardiac artery aneurysms or stenosis (e.g., without an initial biomarker-based Kawasaki disease diagnosis). This method includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) biomarkers of Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17 in a biological sample obtained from the subject. According to this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of at least one protein of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of at least one protein of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative of a subject having a predisposition to develop cardiac artery aneurysms or stenosis.


In a third aspect, the invention features a method for classifying a subject. Such classification includes predicting the response to a Kawasaki disease therapy in a subject, selecting a subject that may benefit from a Kawasaki disease therapy, selecting a subject who may benefit from IVIG therapy, or predicting the responsiveness of a subject to IVIG therapy. This method includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) proteins of Table 1 and/or Table 2 in a biological sample obtained from the subject and/or determining whether IgG in the sample binds to one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, forty, fifty, sixty, or more) peptides of Table 3. According to this method, a subject is classified based on at least one or more of the proteins of Table 1 having an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or at least one or more of the proteins of Table 2 having a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of IgG in said sample to a peptide of Table 3, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease).


In some embodiments, the method further includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) additional biomarkers in the biological sample. In certain embodiments, the one or more additional proteins are any protein of Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9; an m RNA of Table 10, Table 11, Table 12, and/or Table 13; and/or a glycan of Table 14, Table 15, Table 16, and/or Table 17. By this step a subject can be further classified (for example, by determining the likelihood of a subject to develop cardiac artery aneurysms or stenosis, predicting the response to a Kawasaki disease therapy, selecting a subject that may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy, or predicting the responsiveness of a subject to IVIG therapy) based on one or more of the proteins of Table 4, Table 6, and/or Table 8, one or more mRNA of Table 10 or Table 12, and/or one or more glycan of Table 14 or Table 16 having an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) and/or one or more of the proteins of Table 5, Table 7, and/or Table 9, one or more mRNA of Table 11 or Table 13, and/or one or more glycan of Table 15 or Table 17 having a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment).


In a fourth aspect, the invention features a method for classifying a subject. Such classification includes determining the likelihood of a subject to develop cardiac artery aneurysms or stenosis, predicting the response to a Kawasaki disease therapy, selecting a subject that may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy, or predicting the responsiveness of a subject to IVIG therapy. The method includes: determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) biomarkers of Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17 in a biological sample. According to this method, a subject is classified based on at least one or more of the Table 4, Table 6, and/or Table 8, one or more mRNA of Table 10 or Table 12, and/or one or more glycan of Table 14 or Table 16 having an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more), as compared to a reference (e.g., a control, such as, a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) and/or one or more Table 5, Table 7, and/or Table 9, one or more mRNA of Table 11 or Table 13, and/or one or more glycan of Table 15 or Table 17 having a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment).


In a fifth aspect, the invention features a method for treating Kawasaki disease. The method includes: (a) determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) proteins from Table 1 or Table 2 in a biological sample obtained from the subject and/or determining whether IgG in the sample binds to one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, forty, fifty, sixty, or more) peptides of Table 3; and (b) administering a Kawasaki disease therapy to the subject if the level of the one or more proteins is indicative that the subject may benefit from a Kawasaki disease therapy (e.g., administration of IVIG). In this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 1, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy, and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 2, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of IgG in said sample to a peptide of Table 3, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative that the subject may benefit from a Kawasaki disease therapy.


In some embodiments, the method further includes, prior to the determining step, the step of selecting a subject having a fever and one or more of: red eyes; a red swollen tongue; red skin on the palms on the hands and/or soles of the feet; peeling skin on the hands and/or feet; a rash on the main part of the body and/or in the genital area; and swollen lymph nodes and/or the step of obtaining a biological sample from said subject.


In other embodiments, the method further includes, between step (a) and step (b), the step of comparing the level of said one or more proteins to a reference (e.g., a predetermined control value) and/or comparing the binding of IgG in the sample to a reference (e.g., a predetermined control value).


In some embodiments, the method further includes (c) determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) biomarkers from Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17 in a biological sample obtained from the subject; and (d) administering a Kawasaki disease therapy other than, or in addition to, IVIG therapy (e.g., a Kawasaki disease therapy including one or more anticoagulants such as enoxaparin and/or clopidogrel, an anti-inflammatory such as aspirin, and/or one or more immunosuppressant drugs such as infliximab, cyclosporine, and/or prednisone) to the subject if the level of the one or more biomarkers is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy. In this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy (e.g., a Kawasaki disease therapy including one or more anticoagulants such as enoxaparin and/or clopidogrel, an anti-inflammatory such as aspirin, and/or one or more immunosuppressant drugs such as infliximab, cyclosporine, and/or prednisone); and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


In a sixth aspect, the invention features a method of treating Kawasaki disease. The method includes: (a) determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) biomarkers from Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17 in a biological sample obtained from the subject; and (b) administering a Kawasaki disease therapy other than, or in addition to, IVIG therapy (e.g., a Kawasaki disease therapy including one or more anticoagulants such as enoxaparin and/or clopidogrel, an anti-inflammatory such as aspirin, and/or one or more immunosuppressant drugs such as infliximab, cyclosporine, and/or prednisone) to the subject if the level of the one or more biomarkers is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy. In this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy (e.g., a Kawasaki disease therapy including one or more anticoagulants such as enoxaparin and/or clopidogrel, an anti-inflammatory such as aspirin, and/or one or more immunosuppressant drugs such as infliximab, cyclosporine, and/or prednisone); and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


Any of the methods herein that rely upon protein measurement can also be adapted for use with the measurement of mRNA levels for the protein. Accordingly, in a seventh aspect, the invention features a method for diagnosing Kawasaki disease in a subject, diagnosing whether a subject has a predisposition to develop cardiac artery aneurysms or stenosis, classifying a subject, or treating Kawasaki disease. This method includes the step of determining the level of mRNA encoding one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) proteins of Tables 1-9 in a biological sample obtained from the subject. According to this method, increased level of mRNA (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) encoding a protein of Table 1, Table 4, Table 6, and/or Table 8 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is a basis for classification of the subject and/or is indicative of the subject having Kawasaki disease, of the subject having a predisposition to develop cardiac artery aneurysms or stenosis, that the subject may benefit from a Kawasaki disease therapy, or that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy; and/or decreased mRNA level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 2, Table 5, Table 7, and/or Table 9 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is a basis for classification of the subject and/or is indicative of the subject having Kawasaki disease, of the subject having a predisposition to develop cardiac artery aneurysms or stenosis, that the subject may benefit from a Kawasaki disease therapy, or that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


In some embodiments, the method further includes the step of administering a Kawasaki disease therapy or a Kawasaki disease therapy other than, or in addition to, IVIG therapy if the mRNA level of the one or more proteins is indicative that the subject may benefit from a Kawasaki disease therapy or a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


In other embodiments, the method further includes prior to determining the expression level, extracting mRNA from the biological sample and reverse transcribing the mRNA into cDNA to obtain a treated biological sample.


In certain embodiments, the mRNA level is determined by an amplification-based assay (e.g., PCR, quantitative PCR, or real-time quantitative PCR), amplification-free assay (e.g., Nanostring), microdroplet based assay, nanopore based assay, or bead based assays (e.g., Luminex, nanoparticles, Nanosphere).


Next generation sequencing methods may also be used with the methods of the invention. Next generation sequencing methods are sequencing technologies that parallelize the sequencing process, producing thousands or millions of sequences concurrently (see, for example, Hall, J. Exp. Biol. 209(Pt.9):1518-1525 (2007) for a review of next generation methods). Next generation sequencing methods include, but are not limited to, polony sequencing, 454 pyrosequencing, Illumina (Solexa) sequencing, SOLiD sequencing, Ion Torrent semiconductor sequencing, DNA nanoball sequencing, Heliscope single molecule sequencing, single molecule real time sequencing, nanopore DNA sequencing (see, for example, Dela Torre et al. Nanotechnology, 23(38):385308, 2012), tunneling currents DNA sequencing (see, for example, Massimiliano, Nanotechnology, 24:342501, 2013), sequencing by hybridization (see, for example, Qin et al. PLoS One, 7(5):e35819, 2012), sequencing with mass spectrometry (see, for example, Edwards et al. Mutation Research, 573(1-2):3-12, 2005), microfluidic Sanger sequencing (see, for example, Kan et al. Electrophoresis, 25(21-22):3564-3588, 2004), microscopy-based sequencing (see, for example, Bell et al. Microscopy and microanalysis: the official journal of Microscopy Society of America, Microbeam Analysis Society, Microscopical Society of Canada, 18(5):1-5, 2012), and RNA polymerase sequencing (see, for example, Pareek et al. J. Applied Genetics, 52(4):413-415, 2011).


In an eighth aspect, the invention features a kit or device for selecting a subject that may benefit from a Kawasaki disease therapy. The kit or device includes a set of two or more (e.g., three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) distinct binding agents, each of the binding agents being capable of specifically binding to at least one protein from Table 1 and/or Table 2, wherein each binding agent binds a different protein and/or one or more peptides from Table 3.


In a ninth aspect, the invention features a kit or device for selecting a subject that may benefit from a Kawasaki disease therapy. The kit or device includes a set of two or more (e.g., three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) distinct reagents, each or the reagents being capable of detecting at least one mRNA that encodes a protein from Table 1 and/or Table 2, wherein each reagent detects a different mRNA that encodes a protein from Table 1 and/or Table 2.


Optionally the kit or device also includes instructions for use of the kit or device to determine the level of the proteins in a biological sample and/or instructions for use of the kit or device to determine the binding of IgG in the sample to the one or more peptides of Table 3.


In some embodiments of any of the foregoing kits or devices, the kit or device further includes a set of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) binding agents, each of the binding agents being capable of specifically binding to at least one protein, or the mRNA which encodes the protein, from Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9; an mRNA or protein product of an mRNA of Table 10, Table 11, Table 12, and/or Table 13; and/or a glycan of Table 14, Table 15, Table 16, and/or 17.


In a tenth aspect, the invention features a kit or device for selecting a subject that may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy. The kit or device includes a set of two or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) binding agents, each of the binding agents being capable of specifically binding to at least one protein from Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9; an mRNA or protein product of an mRNA of Table 10, Table 11, Table 12, and/or Table 13; and/or a glycan of Table 14, Table 15, Table 16, and/or 17 wherein each binding agent binds a different biomarker.


In an eleventh aspect, the invention features a kit or device for selecting a subject that may benefit from a Kawasaki disease therapy. The kit or device includes a set of two or more (e.g., three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) distinct reagents, each or the reagents being capable of detecting at least one mRNA that encodes a protein from Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9 wherein each reagent detects a different mRNA that encodes a protein from Table 4, Table 5, Table 6, Table 7, Table 8, and/or Table 9.


In a twelfth aspect, the invention features a method for diagnosing Kawasaki disease in a subject. The method includes determining the level of one or more proteins in a biological sample obtained from the subject with any of the foregoing kits or devices, wherein an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 1, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 2, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of IgG in said sample to a peptide of Table 3, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative of the subject having Kawasaki disease.


In some embodiments, the method further includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) additional biomarkers in the biological sample. In certain embodiments, the one or more additional biomarkers are any biomarker of Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17. A subject can be further diagnosed with a predisposition to develop cardiac artery aneurysms or stenosis based on one or more of the proteins of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16 having an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) and/or one or more of the proteins of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17 having a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less), as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease or a subject that has Kawasaki disease and responded positively to IVIG treatment) is indicative of the subject having a predisposition to develop cardiac artery aneurysms or stenosis.


In a thirteenth aspect, the invention features a method for diagnosing whether a subject has a predisposition to develop cardiac artery aneurysms or stenosis. This method includes the step of determining the level of one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, twelve, fifteen, twenty, twenty-five, thirty, or more) biomarkers of Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17 in a biological sample obtained from the subject with any of the foregoing kits or devices. According to this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of at least one protein of Table 4, Table 6 and/or Table 8, and/or at least one glycan of Table 11, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of at least one protein of Table 5, Table 7, and/or Table 9, at least one mRNA of Table 11 or Table 13, and/or at least one glycan of Table 15 or Table 17, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative of a predisposition to develop cardiac artery aneurysms or stenosis in said subject.


In a fourteenth aspect, the invention features a method for treating Kawasaki disease in a subject. The method includes the steps of (a) determining the level of one or more proteins in a biological sample obtained from the subject and/or binding of IgG in the sample to one or more peptides with any of the foregoing kits or devices; and (b) administering a Kawasaki disease therapy to the subject if the level of the one or more proteins is indicative that the subject may benefit from a Kawasaki disease therapy (e.g., administration of IVIG). In this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 1, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy, and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 2, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), and/or increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of IgG in said sample to a peptide of Table 3, as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease) is indicative that the subject may benefit from a Kawasaki disease therapy.


In a fifteenth aspect, the invention features a further method for treating Kawasaki disease in a subject. This method includes the steps of (a) determining the level of one or more biomarkers in a biological sample obtained from the subject with any of the foregoing kits or devices; and (b) administering a therapy other than, or in addition to, IVIG therapy to the subject if the level of the one or more proteins is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy. In this method, an increased level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5M-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more) of a protein of Table 4, Table 6, and/or Table 8, an mRNA of Table 10 or Table 12, and/or a glycan of Table 14 or Table 16 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy (e.g., a Kawasaki disease therapy including one or more anticoagulants such as enoxaparin and/or clopidogrel, an anti-inflammatory such as aspirin, and/or one or more immunosuppressant drugs such as infliximab, cyclosporine, and/or prednisone); and/or a decreased level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less) of a protein of Table 5, Table 7, and/or Table 9, an mRNA of Table 11 or Table 13, and/or a glycan of Table 15 or Table 17 as compared to a reference (e.g., a control, such as a predetermined control value, or a sample from a subject that does not have Kawasaki disease), is indicative that the subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


In other embodiments of any of the foregoing methods, the Kawasaki disease therapy includes administration of IVIG to the subject (e.g., in high doses such as greater than 400 mg/kg, 500 mg/kg, 600 mg/kg, 700 mg/kg, 800 mg/kg, 900 mg/kg, 1 g/kg, 1.1 g/kg, 1.2 g/kg, 1.3 g/kg, 1.4 g/kg, 1.5 g/kg, 1.6 g/kg, 1.7 g/kg, 1.8 g/kg, 1.9 g/kg, 2.0 g/kg, 2.10 g/kg, 2.20 g/kg, 2.3 g/kg, 2.4 g/kg, 2.5 g/kg or more). In some embodiments, IVIG is administered to the subject between the fifth and ninth day after the appearance of symptoms. In other embodiments of any of the foregoing methods, the Kawasaki disease therapy includes administration of one or more anticoagulants (e.g., enoxaparin and/or clopidogrel or a pharmaceutically acceptable salt thereof) to the subject. In some embodiments of any of the foregoing methods, the Kawasaki disease therapy includes administration of an anti-inflammatory agent (e.g., aspirin). In other embodiments of any of the foregoing methods, the Kawasaki disease therapy includes administration of one or more immunosuppressant drugs (e.g., infliximab, cyclosporine, and/or prednisone).


In certain embodiments of any of the foregoing methods, the subject has one or more of: a fever; red eyes; a rash on the main part of the body and/or in the genital area; red, dry, cracked lips; a red, swollen tongue; swollen, red skin on the palms of the hands and/or soles of the feet; swollen lymph nodes; irritability; peeling of the skin on the hands and/or feet; joint pain; diarrhea; vomiting; and abdominal pain. For example, the subject may have a fever (e.g., a fever lasting more than four days) and one or more of: red eyes; a red swollen tongue; red skin on the palms of the hands and/or soles of the feet; peeling of the skin on the hands and/or feet; a rash on the main part of the body and/or in the genital area; and swollen lymph nodes.


In other embodiments of any of the foregoing methods, the subject exhibits the clinical symptoms of cardiac artery aneurysms and/or stenosis of the arteries. In yet other embodiments of any of the foregoing methods, the subject has not been diagnosed with cardiac artery aneurysms and/or stenosis of the arteries prior to determining the level of the one or more proteins. In certain embodiments of any of the foregoing methods, the subject has a white blood cell count and/or a C-reactive protein measurement that is not indicative of inflammation.


In some embodiments of any of the foregoing methods, the biological sample is obtained from the subject prior to the commencement of IVIG therapy. In other embodiments of any of the foregoing methods, the biological sample is obtained from the subject after commencement of IVIG therapy. In certain embodiments of any of the foregoing methods, the biological sample is obtained from the subject with 24 hours after commencement of IVIG therapy. In some embodiments of any of the foregoing methods, the biological sample is a tissue sample, whole blood, plasma, urine, saliva, pancreatic juice, bile, or serum sample. In certain embodiments of any of the foregoing methods, the biological sample is a plasma sample.


In some embodiments of any of the foregoing methods, the biological sample is processed prior to determining the level of the one or more the proteins, e.g., the biological sample is centrifuged, the biological sample is filtered, the biological sample is diluted, the biological sample is treated with reagents (e.g., digesting enzymes or reducing reagents), the biological sample is fractionated to remove more abundant proteins (e.g., proteins present at concentrations greater than 0.01 g/dL, greater than 0.02 g/dL, greater than 0.05 g/dL, greater than 0.1 g/dL, greater than 0.2 g/dL, greater than 0.5 g/dL, greater than 1.0 g/dL, greater than 2.0 g/dL, greater than 3.0 g/dL), such as, albumins, globulins (e.g., haptoglobulin, alpha2-macroglobulin, IgG, IgA, and IgM), alpha1-acid glycoprotein, apolipoprotein AI, apolipoprotein AII, complement C3, transthyretin, antitrypsin, transferrin, and fibrinogen and/or enrich for less abundant proteins, such as, any protein from Tables 1, 2, 4, or 5. In some embodiments, the biological sample is subjected to centrifugation to remove red blood cells. In certain embodiments, the biological sample is filtered (e.g., spin filtered). In some embodiments, the biological sample is diluted. In other embodiments, the biological sample is subjected to cold alcohol fractionation. In certain embodiments, the biological sample is subjected to chromatographic separation (e.g., using an immunoaffinity-based column). In some embodiments, the biological sample is concentrated. In other embodiments, the biological sample is buffer exchanged. In certain embodiments, the biological sample is treated with a digesting enzyme (e.g., trypsin).


In other embodiments any of the foregoing methods further include contacting the biological sample with one or more binding agents capable of specifically binding to the one or more proteins, one or more peptides of Table 3, one or more mRNAs of Table 10, Table 11, Table 12, and/or Table 13, and/or one or more glycans of Table 14, Table 15, Table 16, and/or Table 17.


In any of the aspects and embodiments described herein, the protein level and/or binding of IgG in the sample is determined by one or more of a hybridization assay, an immunoassay, liquid chromatography, mass spectrometry, and/or fluorescence in situ hybridization assay (e.g., Northern analysis, ELISA, immunohistochemical analysis, microarray, chip, microfluidic chip, sequencing, or Western blotting).


In certain embodiments of any of the foregoing methods, the subject is less than 18 years old (e.g., less than 17 years old, less than 16 years old, less than 15 years old, less than 14 years old, less than 13 years old, less than 12 years old, less than 11 years old, less than 10 years old, less than 9 years old, less than 8 years old, less than 7 years old, less than 6 years old, less than 5 years old, less than 4 years old, less than 3 years old, less than 2 years old, less than 1 year old, less than 6 months old). In some embodiments of any of the foregoing methods, the subject is Asian (e.g., Japanese or Korean) or Afro-Caribbean.


In other embodiments of any of the foregoing methods, the level of the one or more proteins and/or binding of IgG in the sample is determined at least twice within 365 days (e.g., twice within 180 days, within 90 days, within 60 days, within 30 days, within 14 days, within 7 days). In certain embodiments, the level of the one or more proteins and/or binding of IgG in the sample is determined at least once prior to the commencement of IVIG therapy and at least once after commencement of IVIG therapy.


In certain embodiments any of the foregoing kits or devices also include instructions for use of the kit or device to determine the level of the proteins in a biological sample, the binding of IgG to a peptide of Table 3, the expression level of an mRNA of Table 10, Table 11, Table 12, and/or Table 13, and/or the abundance of a glycan of Table 14, Table 15, Table 16, and/or Table 17.


In other embodiments of any of the foregoing methods, the method further includes the step of recording the result in a print or computer readable media. In other embodiments, the method further includes the step of informing (e.g., providing the results of the determining step on printable media) the subject that he or she has Kawasaki disease, may benefit from a Kawasaki disease therapy, may benefit from IVIG therapy, may have an increased likelihood to develop cardiac artery aneurysms and/or stenosis, may have a predisposition to develop cardiac artery aneurysms and/or stenosis, may benefit from a therapy other than, or in addition to, IVIG therapy, or may benefit from therapy that includes one or more anticoagulants, an anti-inflammatory agent, and/or one or more immunosuppressant drugs.


In some embodiments of any of the aspects described herein, the binding agent is an antibody. In other embodiments of any of the aspects described herein, one or more of the binding agents and/or peptides of Table 3 are provided on a solid support (e.g., as a microarray). In any of the aspects and embodiments described herein, the one or more proteins, one or more peptides of Table 3, one or more mRNAs of Table 10, Table 11, Table 12, and/or Table 13, one or more glycans of Table 14, Table 15, Table 16 and/or Table 17, and/or set of binding agents and/or peptides include or consist of any combination described herein. In any of the aspects and embodiments described herein, the one or more peptides of Table 3 may be attached to a solid support by a linker (e.g., an N-terminal or C-terminal cysteine, or an N-terminal or C-terminal cysteine-serine-glycine group).


Also provided herein are methods of monitoring a subject with Kawasaki disease. The diagnostic kits and methods disclosed herein can be used to determine an optimal treatment plan for a subject or to determine the efficacy of a treatment plan for a subject. For example, the subject can be treated for Kawasaki disease and the prognosis of the disease can be determined by the diagnostic kits and methods disclosed herein. In particular embodiments, a diagnostic kit or method is used to determine if a subject has Kawasaki disease. A diagnostic kit or method can include a screen for protein level and/or IgG binding profiles by any useful detection method (e.g., unlabeled, fluorescence, radiation, or chemiluminescence). A diagnostic test can further include one or more binding agents (e.g., one or more of probes, primers, peptides, small molecules, aptamers, or antibodies) to detect the level of these proteins or mRNAs encoding these proteins. In certain embodiments, the diagnostic kit includes the use of one or more proteins associated with Kawasaki disease and/or one or more peptides of Table 3 in a diagnostic platform, which can be optionally automated.


Also provided herein are general strategies to develop diagnostic tests which can be used to diagnose Kawasaki disease based on the level of proteins, binding of IgG to one or more peptides of Table 3, mRNAs of Table 10, Table 11, Table 12, and/or Table 13, one or more glycans of Table 14, Table 15, Table 16 and/or Table 17 disclosed herein. These strategies can be used to develop tests that use one or more of these proteins, peptides, mRNAs, and/or glycans, any combination of one or more of these proteins, peptides, mRNAs and/or glycans, one or more of these proteins, peptides, mRNAs, and/or glycans in combination with any other biomarkers found to be associated with Kawasaki disease, and/or one or more of these proteins, peptides, mRNAs, and/or glycans in combination with one or more reference biomarkers not associated with Kawasaki disease.


Also provided herein are methods of determining the likelihood of a subject to develop cardiac artery aneurysms or stenosis. Accordingly, the invention also includes methods of diagnosing a subject that would benefit from a therapy other than or in addition to, IVIG therapy by performing any of the methods or using any of the compositions or kits described herein.


Other features and advantages of the invention will be apparent from the following description and the claims.


Definitions

As used herein, the term “about” means ±10% of the recited value.


The term “array” or “microarray,” as used herein refers to an ordered arrangement of hybridizable array elements, preferably protein probes (e.g., antibodies), on a substrate. The substrate can be a solid substrate, such as a glass slide, beads, or microfluidic chip, or a semi-solid substrate, such as nitrocellulose membrane.


The term “Afro-Caribbean” refers to a person of Caribbean descent (i.e., is from or has an ancestor from the Caribbean Region, as classified by the United Nations Department of Economic and Social Affairs) and has an ancestor that emigrated from Africa to the Caribbean Region in the period since 1492.


The term “anticoagulant” refers to a drug that works to prevent the coagulation of blood, such as coumarins, thienopyridines (e.g., clopidogrel), heparin, low molecular weight heparin (e.g., enoxaparin), inhibitors of factor Xa, or thrombin inhibitors.


The term “anti-inflammatory agent” refers to a drug that reduces inflammation in a subject, e.g., non-steroidal anti-inflammatory drugs (NSAIDs) such as aspirin, ibuprofen, and naproxen.


The term “Asian” refers to a person of Asian descent (i.e., is from or has an ancestor from the Eastern Asia or Southeastern Asia Regions, as classified by the United Nations Department of Economic and Social Affairs). For example, a person from, or having an ancestor from, Japan (i.e., someone who is Japanese); or a person from, or having an ancestor from, Korea (i.e., someone who is Korean) are Asian.


By a “binding agent” is meant any compound (e.g., a probe, primer, protein, small molecule, aptamer, or antibody) capable of specifically binding a target. By “specifically binds” is meant binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule. For example, specific binding can be determined by competition with a control molecule that is similar to the target, for example, an excess of non-labeled target. In this case, specific binding is indicated if the binding of the labeled target to a binding agent is competitively inhibited by excess unlabeled target. The term “specific binding,” “specifically binding,” or “specifically binds to” a particular protein as used herein can be exhibited, for example, by a molecule having a KD for the target of 10−4 M or lower, alternatively 10−5 M or lower, alternatively 10−6 M or lower, alternatively 10−7 M or lower, alternatively 10−5 M or lower, alternatively 10−9 M or lower, alternatively 10−10 M or lower, alternatively 10−11 M or lower, alternatively 10−12 M or lower, or a KD in the range of 10−4 M to 10−12 M or 10−6 M to 10−10 M or 10−7 M to 10−9 M. As will be appreciated by the skilled artisan, affinity and KD values are inversely related. A high affinity for a target is measured by a low KD value. In one embodiment, the term “specific binding” refers to binding where a binding agent binds to a particular protein, mRNA, or glycan without substantially binding to any other protein, mRNA, or glycan.


By “biological sample” or “sample” is meant a fluid or solid sample from a subject. Biological samples may include cells; nucleic acid, protein, or membrane extracts of cells; or blood or biological fluids including (e.g., plasma, serum, saliva, urine, bile). Solid biological samples include samples taken from feces, the rectum, central nervous system, bone, breast tissue, renal tissue, the uterine cervix, the endometrium, the head or neck, the gallbladder, parotid tissue, the prostate, the brain, the pituitary gland, kidney tissue, muscle, the esophagus, the stomach, the small intestine, the colon, the liver, the spleen, the pancreas, thyroid tissue, heart tissue, lung tissue, the bladder, adipose tissue, lymph node tissue, the uterus, ovarian tissue, adrenal tissue, testis tissue, the tonsils, and the thymus. Fluid biological samples include samples taken from the blood, serum, plasma, pancreatic fluid, CSF, semen, prostate fluid, seminal fluid, urine, saliva, sputum, mucus, bone marrow, lymph, and tears. Samples may be obtained by standard methods including, e.g., venous puncture and surgical biopsy. In certain embodiments, the biological sample is a blood, plasma, or serum sample.


By “classifying a subject” is meant predicting a response to a Kawasaki disease therapy by a subject; selecting a subject that may benefit from a Kawasaki disease therapy; selecting a subject who may benefit from IVIG therapy; predicting the responsiveness of a subject to IVIG therapy; determining the likelihood of a subject to develop cardiac artery aneurysms or stenosis; or selecting a subject that may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


By “diagnosing” is meant identifying a molecular or pathological state, disease or condition, such as the identification of Kawasaki disease or cardiac artery aneurysm and/or stenosis, or to refer to identification of a subject having Kawasaki disease who may benefit from a particular treatment regimen.


By “determining the level of a protein, mRNA, or glycan” is meant the detection of a protein, mRNA, or glycan by methods known in the art either directly or indirectly. “Directly determining” means performing a process (e.g., performing an assay or test on a sample or “analyzing a sample” as that term is defined herein) to obtain the physical entity or value. “Indirectly determining” refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value). Methods to measure protein level generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, liquid chromatography (LC)-mass spectrometry, microcytometry, microscopy, fluorescence activated cell sorting (FACS), and flow cytometry, as well as assays based on a property of a protein including, but not limited to, enzymatic activity or interaction with other protein partners. Methods to measure mRNA and glycan levels are known in the art. Exemplary methods are provided herein.


By “determining the binding of IgG” is meant the detection of binding of IgG in a sample (e.g., a plasma sample) to a binding agent (e.g., a peptide of Table 3) by methods known in the art. “Directly determining” means performing a process (e.g., performing an assay or test on a sample or “analyzing a sample” as that term is defined herein) to obtain the physical entity or value. “Indirectly determining” refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value). Methods to measure binding generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, liquid chromatography (LC)-mass spectrometry, microcytometry, microscopy, fluorescence activated cell sorting (FACS), flow cytometry, peptide arrays, protein arrays and microarrays.


The term “immunosuppressant drug,” as used herein refers to a drug that inhibits or prevents activity of the immune system including glucocorticoids such as prednisone, cytostatics such as methotrexate, antibodies such as infliximab, drugs acting on immunophilins such as cyclosporine.


By “informing a subject” is meant providing the subject or the parent or legal guardian of the subject the results of the determining step and/or analysis of the results verbally and/or on printable media.


The term “IVIG” as used herein refers to intravenous immunoglobulin, a blood product containing pooled, polyvalent IgG extracted from the plasma of over one thousand blood donors. The term “IVIG therapy” refers to a treatment including the administration of IVIG to a subject, e.g., in high doses, such as, 2 g/kg.


By “Kawasaki disease therapy” is meant any therapy in the art for the treatment of Kawasaki disease, such as, therapeutic agents or modalities for Kawasaki disease. Common treatments for Kawasaki disease include administration of IVIG (i.e., IVIG therapy); salicylates (e.g., aspirin); corticosteroids (e.g., prednisone); IL-1 receptor antagonists; anticoagulants (e.g., enoxaparin and/or clopidogrel); anti-TNF agents (e.g., infliximab); or any combination thereof.


The terms “kit or device,” as used herein, refer to a set of articles and/or equipment, such as reagents, instruments, and systems, intended for use in diagnosis or prognosis of disease or other conditions, including determination of the state of health, in order to cure, mitigate, treat, or prevent disease or its sequelae. The kits and devices of the invention are intended for use in the collection, preparation, and/or examination of biological samples taken from the subject. For example, the kits and devices of the invention may be used for biochemical estimation or the qualitative detection of a protein. The kits and devices of the invention may include general purpose reagents and analyte specific reagents. A “general purpose reagent” refers to a chemical reagent that has general laboratory application, used to collect, prepare, and/or examine specimens from the human body for diagnostic purposes, and is not labeled or otherwise intended for a specific diagnostic application. An “analyte specific reagent” refers to antibodies, both polyclonal and monoclonal, specific receptor proteins, ligands, nucleic acids, and other binding agents which, through specific binding or chemical reaction with substances in a biological sample, are intended for use in a diagnostic application for identification and quantification of an individual chemical substance or ligand in biological samples. The kits and devices of the invention may include a label which states the name of the kit or device, the intended use or uses of the device (e.g., the diagnosis of Kawasaki disease), a statement of warnings or precautions for users of any hazardous substances contained in the kit or device and any other warnings appropriate to user hazards, the established name of the reagents, quantity, proportion, or concentration of all active ingredients and for reagents derived from biological activity, the source and measure of its activity, storage instructions, and/or instructions for manipulation of products requiring mixing or reconstitution. The kit may also include instructions for detection read out and interpretation.


By “level” is meant a level of a protein, glycan, or mRNA, as compared to a reference. The reference can be any useful reference, as defined herein. By a “decreased level” or an “increased level” of a protein is meant a decrease or increase in protein level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about 1.2-fold, about 1.4-fold, about 1.5-fold, about 1.8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more). A level of a protein may be expressed in mass/vol (e.g., g/dL, mg/mL, μg/mL, ng/mL) or percentage relative to total protein, glycan, or mRNA in a sample. By “protein level profile” is meant one or more protein level values determined for a sample.


By “processing a sample” is meant any process carried out on the sample prior to the determination of the level or expression of the protein. Exemplary processing steps include, but are not limited to, centrifugation of the sample, fractionation of the sample, treatment with reagents (e.g., digesting enzymes or reducing reagents), and/or dilution of the sample. By “fractionation of a sample” is meant the general processes of separating the various components of a sample. For example, the components of the sample may be separated by chromatography (e.g., ion exchange chromatography). In some cases, the most abundant proteins, such as, proteins present at greater than 0.01, greater than 0.02, greater than 0.05, greater than 0.1 g/dL (e.g., greater than 0.2 g/dL, greater than 0.5 g/dL, greater than 1.0 g/dL, greater than 2.0 g/dL, greater than 3.0 g/dL) are depleted from the sample by chromatography to enhance the sensitivity for less abundant proteins, such as, proteins present at less than 0.2 g/dL (e.g., less than 0.1 g/dL, less than 0.05 g/dL, less than 0.01 g/dL). Columns/kits for the depletion of abundant proteins are known in the art, for example, MARS Human-6 and Human-7 from Agilent Technologies deplete the 6 and 7 most abundant proteins from human plasma.


By “reagent” is meant a polynucleotide sequence or polypeptide sequence capable of detecting a target sequence, or a fragment thereof.


By a “reference” is meant any useful reference used to compare protein or mRNA levels related to Kawasaki disease and/or binding of IgG to a peptide of Table 3. The reference can be any sample, standard, standard curve, or level that is used for comparison purposes. The reference can be a normal reference sample or a reference standard or level. A “reference sample” can be, for example, a control, e.g., a predetermined negative control value such as a “normal control” or a prior sample taken from the same subject; a sample from a normal healthy subject, such as a normal cell or normal tissue; a sample (e.g., a cell or tissue) from a subject not having Kawasaki disease; a sample from a subject that is diagnosed with cardiac artery aneurysms or stenosis; a sample from a subject that has been treated for Kawasaki disease; or a sample of a purified protein (e.g., any described herein) at a known normal concentration. By “reference standard or level” is meant a value or number derived from a reference sample. A “normal control value” is a pre-determined value indicative of non-disease state, e.g., a value expected in a healthy control subject. Typically, a normal control value is expressed as a range (“between X and Y”), a high threshold (“no higher than X”), or a low threshold (“no lower than X”). A subject having a measured value within the normal control value for a particular biomarker is typically referred to as “within normal limits” for that biomarker. A normal reference standard or level can be a value or number derived from a normal subject not having Kawasaki disease; a subject that is diagnosed with cardiac artery aneurysms or stenosis; a subject that has been treated for Kawasaki disease. In preferred embodiments, the reference sample, standard, or level is matched to the sample subject sample by at least one of the following criteria: age, weight, sex, disease stage, and overall health. A standard curve of levels of a purified protein, e.g., any described herein, within the normal reference range can also be used as a reference.


“Response” as used herein indicates a subject's response to a Kawasaki disease therapy, e.g., a response can be a positive response such that symptoms will be alleviated as a result of the Kawasaki disease therapy.


By “selecting a subject” is meant to choose a subject directly or indirectly in preference to others based on an analysis, e.g., analysis of results of the methods of the invention or clinical evaluation. Directly selecting means performing a process (e.g., performing an analysis) to choose a subject. Indirectly selecting refers to receiving the results of an analysis from another party or source (e.g., a third party laboratory that directly performed the analysis).


By “solid support” is meant a structure capable of storing, binding, or attaching one or more binding agents.


By “subject” is meant a human (e.g., a child less than 18 years old, less than 13 years old, less than 8 years old, less than 5 years old, less than 4 years old, less than 3 years old, less than 2 years old, or less than 1 year old). A subject to be treated with a pharmaceutical composition described herein may be one who has been diagnosed by a medical practitioner as having such a disease or condition (e.g., Kawasaki disease) or one at risk for developing a disease or condition (e.g., cardiac artery aneurysm or stenosis).


By “target sequence” is meant a portion of a gene or a gene product, including the mRNA and related cDNA.


By “therapeutic agent” is meant any agent that produces a healing, curative, stabilizing, or ameliorative effect.


A “therapeutically effective amount” of a compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. A therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A therapeutically effective amount also encompasses an amount sufficient to confer benefit, e.g., clinical benefit.


By “treating” is meant administering a composition (e.g., a pharmaceutical composition) for therapeutic purposes or administering treatment to a subject already having a condition or disorder to improve the subject's condition or to reduce the likelihood of a condition or disorder. By “treating a condition or disorder” is meant that the condition or disorder and/or the symptoms associated with the condition or disorder are, e.g., alleviated, reduced, cured, or placed in a state of remission. By “reduce the likelihood of” is meant reducing the severity, the frequency, and/or the duration of a disorder (e.g., cardiac artery aneurysms and/or stenosis) or symptoms thereof. Reducing the likelihood of cardiac artery aneurysms and/or stenosis is synonymous with prophylaxis or the chronic treatment of cardiac artery aneurysms and/or stenosis.


Other features and advantages of the invention will be apparent from the following Detailed Description and the claims.







DETAILED DESCRIPTION OF THE INVENTION

There is no specific test available to diagnose Kawasaki disease. Diagnosis largely is a process of ruling out diseases that cause similar signs and symptoms, including: scarlet fever; juvenile rheumatoid arthritis; Stevens-Johnson syndrome; toxic shock syndrome; measles; certain tick-borne illnesses. A doctor may do a physical examination and perform other tests to help in the diagnosis or prognosis. These tests may include urine tests, blood tests, electrocardiogram, and echocardiogram. (http://www.mayoclinic.org/diseases-conditions/kawasaki-disease/basics/tests-diagnosis/con-20024663, Mar. 3, 2014).


The present invention relates to the identification of biomarkers (e.g., protein levels, mRNA levels, glycan abundance, or IgG binding) that identify subjects having Kawasaki disease and/or being predisposed to develop Kawasaki disease-related cardiac artery aneurysms or stenosis. Such differential levels of proteins, mRNAs, glycans, and/or differential binding of IgG in samples can be used to diagnose, prognose, and classify subjects with Kawasaki disease and/or a predisposition to develop cardiac artery aneurysms or stenosis from healthy controls. Accordingly, the kits and methods described herein are useful for treating or diagnosing Kawasaki disease and/or related cardiac artery aneurysms or stenosis. Also described herein are diagnostic kits (e.g., on a solid support, such as an array or chip) which can be used to perform such methods.


Proteins

Applicants have discovered that the levels of certain proteins can be utilized to diagnose, prognose, and treat Kawasaki disease, as well as to select subjects who would benefit from either IVIG therapy or a Kawasaki disease therapy other than, or in addition to, IVIG therapy. Proteins, the levels of which are of interest in the methods and compositions of the invention, include those in Table 18.









TABLE 18







Proteins with levels relevant to Kawasaki disease









Protein Name
Accession No.
Gene Symbol





collagen, type VI, alpha 3
E7ENL6
COL6A3


F-box protein 47
Q5MNV8
FBXO47


collagen, type VI, alpha 1
P12109
COL6A1


androgen receptor
P10275
AR


tumor protein p53 binding protein 1
Q12888
TP53BP1


peptidase inhibitor 16
Q6UXB8
PI16


cadherin 13
P55290
CDH13


tenascin XB
G5E9A9
TNXB


tenascin XB
B0UYX3
TNXB


peroxisome proliferator-activated
Q07869
PPARA


receptor alpha




collagen, type I, alpha 1
P02452
COL1A1


Ig alpha-2 chain C region (A2m
P01877
IGHA2


marker)




Ig heavy chain V-III region TIL
P01765
HV304


Ig alpha-1 chain C region
P01876
IGHA1


olfactomedin 1
Q6IMJ5
OLFM1


complement factor H
Q5TFM2
CFH


prostaglandin D2 synthase 21 kDa
P41222
PTGDS


(brain)




sex hormone-binding globulin
I3L145
SHBG


Ig kappa chain V-I region
P01593
KV101


gelsolin
P06396
GSN


Ig lambda-1 chain C regions (Mcg
P0CG04
IGLC1


marker)




BMP2 inducible kinase
Q9NSY1
BMP2K


serpin peptidase inhibitor, clade C
P01008
SERPINC1


(antithrombin), member 1




pregnancy-zone protein
P20742
PZP


inter-alpha-trypsin inhibitor heavy
P19827
ITIH1


chain 1




Ig kappa chain V-II region
P01617
KV204


inter-alpha-trypsin inhibitor heavy
P19823
ITIH2


chain 2




complement factor H
P08603
CFH


Hepatocyte growth factor activator
Q04756
HGFAC


attractin
O75882
ATRN


kininogen 1
P01042
KNG1


coagulation factor XIII, B
P05160
F13B


polypeptide




complement factor B
B4E1Z4
CFB


complement component 1, r
H0YFH3
C1R


subcomponent




complement component 1, r
P00736
C1R


subcomponent




extracellular matrix protein 1
Q16610
ECM1


complement component 1, s
P09871
C1S


subcomponent




kininogen 1
C9JEX1
KNG1


complement component 6
P13671
C6


vitamin D binding protein (group-
P02774
GC


specific component)




Alpha-1-antichymotrypsin (serpin
P01011
SERPINA3


peptidase inhibitor, clade A,




member 3)




lactate dehydrogenase C
F5H5G7
LDHC


Alpha-1-antichymotrypsin (serpin
G3V3A0
SERPINA3


peptidase inhibitor, clade A,




member 3)




armadillo repeat containing 2
Q8NEN0
ARMC2


unc-45 homolog A (C. elegans)
Q9H3U1
UNC45A


defensin, alpha 1
P59665
DEFA1


S100 calcium binding protein A9
P06702
S100A9


solute carrier family 26 (anion
P40879
SLC26A3


exchanger), member 3




zeta-chain (TCR) associated protein
P43403
ZAP70


kinase 70 kDa




Zinc finger protein 106 homolog
Q9H2Y7
ZFP106


apolipoprotein B
P04114
APOB


Zinc finger protein 161 homolog
J3QLI2
ZFP161


apolipoprotein M
O95445
APOM


apolipoprotein C-II
P02655
APOC2


apolipoprotein F
F5GXS5
APOF


zinc finger protein 578
I3L1Y6
ZNF578


xin actin-binding repeat containing 2
A4UGR9
XIRP2


lipoprotein, Lp(a)
P08519
LPA


armadillo repeat containing 10
Q8N2F6
ARMC10


olfactory receptor, family 5,
Q8NGL3
OR5D14


subfamily D, member 14




orosomucoid 1
P02763
ORM1


Alpha-1-antitrypsin (serpin
P01009
SERPINA1


peptidase inhibitor, clade A,




member 1)




nitric oxide synthase 2, inducible
4843
NOS2


(Gene)




carcinoembryonic antigen-related
1088
CEACAM8


cell adhesion molecule 8 (Gene)




interleukin 33 (Gene)
90865
IL33


interleukin 6 (interferon, beta 2)
3569
IL6


(Gene)




Plasminogen
P00747
PLG


Vitamin K-dependent protein C
P04070
PROC


Coagulation factor XI
P03951
F11


Apolipoprotein F
F5GXS5
APOF


CD44 antigen
E7EPC6
CD44


Ankyrin repeat domain-containing
Q9UPS8
ANKRD26


protein 26




Lysosome-associated membrane
B4E2S7
LAMP2


glycoprotein 2




Basal cell adhesion molecule
P50895
BCAM


Multimerin-1
Q13201
MMRN1


Transforming growth factor-beta-
G8JLA8
TGFBI


induced protein ig-h3




Methylcytosine dioxygenase TET2
E7EQS8
TET2


Alpha-1-antichymotrypsin
P01011
SERPINA3


Cystatin-C
P01034
CST3


matrix metallopeptidase 1
4312
MMP1


(interstitial collagenase) (Gene)




Aftiphilin
Q6uLP2
AFTPH


Death domain containing 1
Q6ZMT9
DTHD1


Metastasis associated 1 family,
E7EV10
MTA


member 3




HGF activator
Q04756
HGFAC


Bone marrow stromal cell antigen 1
Q10588
BST1


Mannan-binding lectin serine
O00187
MASP2


peptidase 2




HCG2014417, isoform CRA a
B7Z718
AGAP2


T-Iymphoma invasion and
F5GZ53
TIAM1


metastasis-inducing protein 1




Hemoglobin subunit gamma-1
P69891
HBG1


Hemoglobin subunit gamma-2
P69892
HBG2


Protein S100-A9
P06702
S100A9


Protein S100-A8
P05109
S100A8


Leucine-rich alpha-2-glycoprotein
P02750
LRG1


Actin, cytoplasmic 1
P60709
ACTB


Haptoglobin-related protein
P00739
HPR


Cathepsin D
P07339
CTSD


C-reactive protein
P02741
CRP


Kallistatin
P29622
SERPINA4


Kininogen-1
C9JEX1
KNG1


Lumican
P51884
LUM


Alpha-2-HS-glycoprotein
P02765
AHSG


Butyrylcholinesterase
P06276
BCHE


Coagulation factor IX
P00740
F9


Ig heavy chain V-III region BRO
P01766
HV305


Selenoprotein P
P49908
SEPP1


Cadherin-13
P55290
CDH13


Myosin-8
P13535
MYH8


Orosomucoid 2
P19652
ORM2


Polycystin (PKD) family receptor for
Q9NTG1
PKDREJ


egg jelly




Complement component 9
P02748
C9


Complement component 2
P06681
C2


Complement factor H-related 3
Q6NSD3
CFHR3


Clusterin
P10909
CLU


Kallikrein B, plasma (Fletcher
H0YAC1
KLKB1


factor) 1




Macrophage stimulating 1
P26927
MST1


(hepatocyte growth factor-like)




Biotinidase
F8W1Q3
BTD


Peptidoglycan recognition protein 2
Q96PD5
PGLYRP2


Fibulin 1
B1AHL2
FBLN1


fibulin 1
P23142
FBLN1


Mannan-binding lectin serine
P48740
MASP1


peptidase 1 (C4/C2 activating




component of Ra-reactive factor)




Alpha-1-microglobulin/bikunin
P02760
AMBP


precursor




Mannan-binding lectin serine
F8W876
MASP1


peptidase 1 (C4/C2 activating




component of Ra-reactive factor)




Apolipoprotein H (beta-2-
P02749
APOH


glycoprotein I)




TBC1 domain family, member 8B
J3KN75
TBC1D8B


(with GRAM domain)




Afamin
P43652
AFM


Heat shock 70 kDa protein 5
P11021
HSPA5


(glucose-regulated protein, 78 kDa)




IgA
NA
IGA


ST6 beta-galactosamide alpha-2,6-
NA
ST6


sialyltranferase 1




haptoglobin
P00738
HP


Matrix metallopeptidase 9
P14780
MMP9


Nidogen-1
P14543
NID1


Neural cell adhesion molecule L1-
O00533
CHL1


like protein




Coagulation factor X
P00742
F10


Titin
Q8WZ42
TTN


Insulin-like growth factor binding
P35858
IGFALS


protein, acid labile subunit




Glycosylphosphatidylinositol
P80108
GPLD1


specific phospholipase D1




Retinol binding protein 4, plasma
Q5VY30
RBP4


Centrosomal protein 70 kDa
Q8NHQ1
CEP70


Thrombospondin 4
P35433
THBS4


Fibronectin 1
F8W7G7
FN1


Insulin-like growth factor 2
P01344
IGF2


(somatomedin A)




Neuropilin 1
O14786
NRP1


Neural cell adhesion molecule 1
E9PLH7
NCAM1


alpha-1-B glycoprotein
P04217
A1BG


Coagulation factor XIII, A1
P00488
F13A1


polypeptide




Apolipoprotein C-1
P02654
APOC1


Carboxypeptidase
Q96IY4
CPB2


Zinc finger protein 217
O75362
ZNF217


Collagen, type XI, alpha 2
H0YHY3
COL11A2


Selectin L
P14151
SELL


Ectonucleotide pyrophosphatase/
Q13822
ENPP2


phosphodiesterase 2




Coagulation factor 11 (thrombin)
P00734
F2


Apolipoprotein D
P05090
APOD


Insulin growth factor
P17936
IGFBP3


binding protein 3




Dehydrogenase/reductase (SDR
A8MXC2
DHRS11


family) member 11




Quiescin Q6 sulfhydryl oxidase 1
O00391
QSOX1


Lipopolysaccharide binding protein
P18428
LBP


Hemaglobin, alpha 1
P69905
HBA1


Hemaglobin, delta
P02042
HBD


Fc fragment of IgG, low affinity IIIa
P08637
FCGR3A


receptor (CD16a)




Hemaglobin, gamma G
P69892
HBG2


Hemaglobin, gamma A
P69891
HBG1


Hemaglobin, beta
P68871
HBB


Kinesin family member 20B
Q96Q89
KIF20B


Lysozyme
P61626
LYZ


Actin, alpha, cardiac muscle 1
P68032
ACTC1


Inter-alpha-trypsin inhibitor heavy
Q06033
ITIH3


chain 3




Inter-alpha-trypsin inhibitor heavy
Q14624
ITIH4


chain family, member 4




Inter-alpha-trypsin inhibitor heavy
B7ZKJ8
ITIH4


chain family, member 4




Inter-alpha-trypsin inhibitor heavy
E9PGN5
ITIH4


chain family, member 4




Actin, beta-like 2
Q562R{grave over ( )}
ACTBL2


Hemaglobin, epsilon 1
P02100
HBE1


Protein phosphatase 6, regulatory
B3KMJ7
PPP6R2


subunit 2




Dedicator of cytokinesis 3
Q8IZD9
DOCK3


Centrosomal protein 290 kDa
O15078
CEP290









As indicated above, proteins useful for diagnosing Kawasaki disease, or selecting or classifying a subject that may benefit from a Kawasaki disease therapy include those in Tables 1 and 2. Proteins useful for selecting or classifying a subject that may benefit from a therapy other than, or in addition to, IVIG therapy include those in Tables 4, 5, 6, and 7.


Applicants have discovered that the level of binding of IgG in samples to certain proteins can be utilized to diagnose, prognose, and treat Kawasaki disease, as well as to select subjects who would benefit from a Kawasaki disease therapy. Proteins, the binding of which are of interest in the methods and compositions of the invention, include those comprising an amino acid sequence of any one of SEQ ID NOs:1 to 68.


Genes

Applicants have discovered that the mRNA expression levels of certain genes can be utilized to diagnose, prognose, and treat Kawasaki disease, as well as to select subjects who would benefit from either IVIG therapy or a Kawasaki disease therapy other than, or in addition to, IVIG therapy. Genes, the mRNA levels of which are of interest in the methods and compositions of the invention, include those in Table 19.









TABLE 19







Genes with mRNA levels relevant to Kawasaki disease











Gene Name
Accession No.
Gene Symbol















CD80 molecule
941
CD80



Matrix metallopeptidase 9





(gelatinase B, 92 kDa gelatinase,
4318
MMP9



92 kDa type IV collagenase)





Colony stimulating factor 2
1439
CSF2RB



receptor, beta, low-affinity





Tumor necrosis factor receptor
7132
TNFRSF1A



superfamily, member 1A





carcinoembryonic antigen-
1088
CEACAM8



related cell adhesion molecule 8





Beta-glucuronidase
2990
GUSB



S100 calcium binding protein
6283
S100A12



A12





Versican
1462
VCAN



Fc fragment of IgG, high affinity
2209
FCGR1A



Ia, receptor (CD64)





S100 calcium binding protein
6282
S100A11



A11





Tumor necrosis factor receptor
7133
TNFRSF1B



superfamily, member 1B





Interleukin 1, beta
3553
IL1B



Interleukin 1 receptor antagonist
3557
IL1RN



S100 calcium binding protein A8
6279
S100A8



Tumor necrosis factor (ligand)
10673
TNFSF13B



superfamily, member 13b





Killer cell lectin-like receptor
10219
KLRG1



subfamily G, member 1





Fc fragment of IgG, high affinity
2212
FCGR2A



IIa, receptor (CD32)





chemokine (C-C motif)
729230
CCR2



receptor 2





Actin, beta
60
ACTB



Tumor necrosis factor
7124
TNF










As indicated above, genes useful for selecting or classifying a subject that may benefit from a therapy other than, or in addition to, IVIG therapy include those in Table 10, Table 11, Table 12, and/or Table 13.


Glycans

Applicants have discovered that the levels of certain glycans on IgG and IgA can be utilized to diagnose, prognose, and treat Kawasaki disease, as well as to select subjects who would benefit from either IVIG therapy or a Kawasaki disease therapy other than, or in addition to, IVIG therapy. Glycans, the levels of which are of interest in the methods and compositions of the invention, include those in Table 20.









TABLE 20







Glycans with levels relevant to Kawasaki disease










Fraction
Structure







GP6HA
FA2[3]G1



GP4HA
A2[6]BG1



GP26HA
FA2BG2S[3,6]2




FA2BG2S[6,6]2



GP11HA
FA2G2



GP5HA
FA2[6]G1



GP7HA
FA2[6]BG1



GP2HA
FA2



GP12HA
FA2BG2



IgG2_G0F_BGIcNAc




IgG2_BGIcNAc




GP3HA
M5




FA2B




A2[6]G1



GP38HA
A4G4S[3,3,3,3]4



GP23HA
A2G2S[3,6]2



IgG2_G1F_BGIcNAc




GP13HA
A2[3]BG1S[3]1




A2[3]BG1S[6]1




M7 D1




FA2[3]G1S[3]1




FA2[3]G1S[6]1



GP10HA
A2BG2



GP41HA
A4G4S[3,3,3,3]4



GP28HA
A3BG3S[3,6]2



IgG3_4_A1
IgG3 isoform




A2F1G1S1



GP37HA
A4F1G3S[3,3,6]3




A4F1G3S[3,6,6]3



GP42HA
A4F1G4S[3,3,3,6]4



GP43HA
A4G4LacS[3,3,3,6]4



GP43LA
A4G4LacS[3,3,3,6]4



GP44HA
A4F3G4S[3,3,3,3]4



GP44LA
A4F3G4S[3,3,3,3]4



GP9HA
A1[3]G1S[3]1




A2G2



GP9LA
A1[3]G1S[3]1




A2G2



GP12LA
FA2BG2




A2[3]BG1S[3]1




A2[3]BG1S[6]1



GP14HA
FA2[3]BG1S[3]1



GP14LA
FA2[3]BG1S[3]1



GP16LA
A2BG2S[6]1



GP18HA
FA2G2S[6]1




FA2BG2S[3]1



GP18LA
FA2G2S[6]1




FA2BG2S[3]1



GP19LA
FA2BG2S[6]1



GP20HA
A2G2S[3,6]2




A3G3S[3]1



GP20LA
A2G2S[3,6]2




A3G3S[3]1



GP24LA
A2BG2S[6,6]2



GP29LA
A4G4S[3]1



GP32HA
A3G3S[3,3,6]3



GP32LA
A3G3S[3,3,6]3



GP34HA
A3G3S[3,3,6]3



GP34LA
A3G3S[3,3,6]3



GP38LA
A4G4S[3,3,3,3]4



GP3LA
M5




FA2B




A2[6]G1



GP41LA
A4G4S[3,3,3,3]4



GP42LA
A4F1G4S[3,3,3,6]4



GP42HA
A4F1G4S[3,3,3,6]4



GP41HA
A4G4S[3,3,3,3]4



GP43LA
A4G4LacS[3,3,3,6]4



GP43HA
A4G4LacS[3,3,3,6]4



GP37HA
A4F1G3S[3,3,3]3




A4F1G3S[3,3,6]3




A4F1G3S[3,6,6]3



GP44HA
A4F3G4S[3,3,3,3]4



GP44LA
A4F3G4S[3,3,3,3]4



GP23HA
A2G2S[3,6]2



GP35HA
A3F1G3S[3,3,3]3



GP31HA
A3G3S[3,3,3]3



IgA_G1_S1
A2,G1,S1



GP13HA
A2[3]BG1S[3]1




A2[3]BG1S[6]1




M7 D1



GP18HA
FA2G2S[6]1



GP12LA
FA2BG2



GP20LA
A2G2S[3,6]2



GP18LA
FA2G2S[6]1



GP12HA
FA2BG2



GP11HA
FA2G2



GP19HA
FA2BG2S[3]1;




FA2BG2S[6]1



GP3HA
M5; FA2B; A2[6]G1



GP10HA
A2BG2



GP5HA
FA2[6]G1



GP6HA
FA2[3]G1



GP13LA
A2[3]BG1S[3]1;




A2[3]BG1S[6]1; M7




D1; FA2[3]G1S[3]1



GP24LA
A2BG2S[6,6]2



GP8HA
FA2[3]BG1; M6 D3



GP7HA
FA2[6]BG1



GP32LA
A3G3S[3,3,6]3



GP26HA
FA2BG2S[3,6]2




FA2BG2S[6,6]2



GP5LA
FA2[6]G1



GP19LA
FA2BG2S[3]1




FA2BG2S[6]1



GP6LA
FA2[3]G1



GP2HA
FA2



GP20HA
A2G2S[3,6]2











G; N-glycans, P; plasma, fraction #, LA; low abundant fraction, HA; high abundant fraction. Glycan annotations according to Oxford Symbol nomenclature. All N-glycans have two core GlcNAcs; F at the start of the abbreviation indicates a core a(1-6)fucose linked to inner GlcNAc; Mx, number (x) of mannose on core GlcNAcs; Ax, number of antenna (GlcNAc) on trimannosyl core; A2, biantennary with both GlcNAcs as b(1-2) linked; A3, triantennary with a GlcNAc linked b(1-2) to both mannose and a third GlcNAc linked b(1,4) to the a(1-3) linked mannose; A3′; triantennary with a GlcNAc linked b(1-2) to both mannose and the third GlcNAc linked b(1-6) mannose; B, bisecting GlcNAc linked b(1-4) to b(1-3) mannose; Gx, number (x) of b1-4 linked galasose on the antenna; Fx, number (x) of linked fucose on antenna, (4) or (3) after the F indicates that the Fuc is a(1-4) or a(1-3) linked to the GlcNAc; Sx, number (x) sialic acids linked to galactose; the number 3 or 6 in parenthesis after S indicates whether the sialic acid is in an a(2-3) or a(2-6) linkage. See Harvey et al. Proposal for a standard system for drawing structural diagrams of N- and O-linked carbohydrates and related compounds. Proteomics 2009, 9:3796-801.


As indicated above, glycans useful for diagnosing Kawasaki disease, selecting or classifying a subject that may benefit from a Kawasaki disease therapy and/or selecting or classifying a subject that may benefit from a therapy other than, or in addition to, IVIG therapy include those in Table 14, Table 15, Table 16, and/or Table 17.


Diagnostic and Classification Methods

The present invention features methods and compositions to diagnose Kawasaki disease. The kits and methods of the invention may be used alone or as a companion diagnostics with other diagnostic or therapeutic approaches, as an early molecular screen to distinguish Kawasaki disease from other diseases and disorders with similar symptoms. More specifically, alterations in the level of one or more proteins described herein (e.g., proteins of Table 1 and/or Table 2) and/or binding of IgG to a protein of Table 3 in a test sample as compared to a normal reference can be used to diagnose Kawasaki disease and/or distinguish Kawasaki disease from diseases or disorders with similar symptoms, thereby allowing subject classification.


Further, the present invention features methods and compositions useful in determining the likelihood of a subject to develop cardiac artery aneurysms and/or stenosis. For example, the methods and compositions of the invention may be used to determine if a subject may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy, by determining the levels of one or more biomarkers of Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15, Table 16, and/or Table 17.


The methods of the invention can be used to diagnose, prognose, or classify a subject, for example, an increase in the level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 1) may indicate a subject has Kawasaki disease and/or may benefit from a Kawasaki disease therapy. Similarly, a decrease in the level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 2) may indicate a subject has Kawasaki disease and/or may benefit from a Kawasaki disease therapy. An increase (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) in binding of IgG in a sample to a protein comprising an amino acid sequence of Table 3 may indicate a subject has Kawasaki disease and/or may benefit from a Kawasaki disease therapy.


Alternatively, an increase in the level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 4) may indicate a subject is predisposed to develop cardiac artery aneurysms or stenosis and/or may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy. Similarly, a decrease in the level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less, as compared to a reference) of a biomarker(s) (e.g., a protein of Table 5) may indicate a subject is predisposed to develop cardiac artery aneurysms or stenosis and/or may benefit from a Kawasaki disease therapy other than, or in addition to, IVIG therapy.


To carry out the methods of the invention, a sample can be obtained by any method known in the art. For instance, samples from a subject may be obtained by venipuncture, resection, bronchoscopy, fine needle aspiration, bronchial brushings, or from sputum, pleural fluid, urine, or blood, such as serum or plasma. Proteins can be detected in these samples. By screening such biological samples, a simple early diagnosis or differential diagnosis can be achieved for Kawasaki disease. In addition, the progress of therapy can be monitored by testing such biological samples for target proteins and/or binding of IgG to a protein of Table 3. Furthermore, the prediction of outcome or response to therapy can similarly be tested using such biological samples for target proteins and/or binding of IgG to a protein of Table 3.


In certain embodiments, the sample may be contacted with an antibody specific for the target protein under conditions sufficient for an antibody-protein complex to form, and detection of the complex. The presence of the biomarker may be detected in a number of ways, such as by Western blotting or ELISA procedures using any of a wide variety of tissues or samples, including plasma or serum. A wide range of immunoassay techniques using such an assay format are available, see, e.g., U.S. Pat. Nos. 4,016,043, 4,424,279, and 4,018,653. These include both single-site and two-site or “sandwich” assays of the noncompetitive types, as well as traditional competitive binding assays. These assays also include direct binding of a labeled antibody to a target biomarker.


Another method involves immobilizing the target biomarkers (e.g., on a solid support) and then exposing the immobilized target to a specific antibody, which may or may not contain a label. Depending on the amount of target and the strength of the label's signal, a bound target may be detectable by direct labeling with the antibody. Alternatively, a second labeled antibody, specific to the first antibody is exposed to the target-first antibody complex to form a target-first antibody-second antibody tertiary complex. The complex is detected by the signal emitted by a label, e.g., an enzyme, a fluorescent label, a chromogenic label, a radionuclide containing molecule (i.e., a radioisotope), or a chemiluminescent molecule.


Variations on the forward assay include a simultaneous assay, in which both sample and labeled antibody are added simultaneously to a bound antibody. These techniques are well known to those skilled in the art, including any minor variations as will be readily apparent. In a typical forward sandwich assay, a first antibody having specificity for the biomarker is either covalently or passively bound to a solid surface (e.g., a glass or a polymer surface, such as those with solid supports in the form of tubes, beads, discs, or microplates), and a second antibody is linked to a label that is used to indicate the binding of the second antibody to the molecular marker.


In alternative methods, the expression of a protein in a sample may be examined using immunohistochemistry (“IHC”) and staining protocols. IHC staining of tissue sections has been shown to be a reliable method of assessing or detecting presence of proteins in a sample. IHC and immunofluorescence techniques use an antibody to probe and visualize cellular antigens in situ, generally by chromogenic or fluorescent methods. The tissue sample may be fixed (i.e., preserved) by conventional methodology (see, e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology,” 3rd edition (1960) Lee G. Luna, HT (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.). One of skill in the art will appreciate that the choice of a fixative is determined by the purpose for which the sample is to be histologically stained or otherwise analyzed. By way of example, neutral buffered formalin, Bouin's or formaldehyde, may be used to fix a sample. Generally, the sample is first fixed and is then dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned. Alternatively, one may section the tissue and fix the sections obtained. The primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety, such as a radioisotope, a colloidal gold particle, a fluorescent label, a chromogenic label, or an enzyme-substrate label.


Alternatively, the levels of biomarkers may be detected without the use of binding agents. In some instances, biological samples as described herein are analyzed, for example, by one or more, enzymatic methods, chromatographic methods, mass spectrometry (MS) methods, chromatographic methods followed by MS, electrophoretic methods, electrophoretic methods followed by MS, nuclear magnetic resonance (NMR) methods, and combinations thereof. In some instances, the biological sample is treated with one or more enzymes (e.g., trypsin). Exemplary chromatographic methods include, but are not limited to, Strong Anion Exchange chromatography using Pulsed Amperometric Detection (SAX-PAD), liquid chromatography (LC), high performance liquid chromatography (HPLC), ultra performance liquid chromatography (U PLC), thin layer chromatography (TLC), amide column chromatography, and combinations thereof. Exemplary mass spectrometry (MS) include, but are not limited to, tandem MS, LC-MS, LC-MS/MS, matrix assisted laser desorption ionisation mass spectrometry (MALDI-MS), Fourier transform mass spectrometry (FTMS), ion mobility separation with mass spectrometry (IMS-MS), electron transfer dissociation (ETD-MS), Multiple Reaction Monitoring (MRM), and combinations thereof. Exemplary electrophoretic methods include, but are not limited to, capillary electrophoresis (CE), CE-MS, gel electrophoresis, agarose gel electrophoresis, acrylamide gel electrophoresis, SDS-polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting using antibodies that recognize specific glycan structures, and combinations thereof. Exemplary nuclear magnetic resonance (NMR) include, but are not limited to, one-dimensional NMR (1 D-NMR), two-dimensional NMR (2D-NMR), correlation spectroscopy magnetic-angle spinning NMR (COSY-NMR), total correlated spectroscopy NMR (TOCSY-NMR), heteronuclear single-quantum coherence NMR (HSQC-NM R), heteronuclear multiple quantum coherence (HMQC-NMR), rotational nuclear overhauser effect spectroscopy NMR (ROESY-NMR), nuclear overhauser effect spectroscopy (NOESY-NMR), and combinations thereof.


Any of the methods herein that rely upon protein measurement can also be adapted for use with the measurement of mRNA levels for the protein. The level of mRNA can be determined using methods known in the art. Methods to measure mRNA levels generally include, but are not limited to, sequencing, northern blotting, RT-PCR, gene array technology, and RNAse protection assays.


Binding Agents


Any binding agent that specifically binds a target biomarker may be used in the methods of the invention. The binding agent may be, e.g., a protein (e.g., an antibody), small molecule, or aptamer capable of specifically binding a target.


Preferably, each binding agent specifically binds to one biomarker in a sample. For determining the level of a biomarker, the measurement of antibodies specific to a biomarker of the invention in a subject may be used for the diagnosis of Kawasaki disease. The binding agent may optionally contain a label, such as a radioisotope, a colloidal gold particle, a fluorescent label, a chromogenic label, an enzyme-substrate label, or a chemiluminescent label.


Sample Processing


In some embodiments of any of the foregoing methods, the biological sample is processed prior to determining the level of the one or more the biomarkers, e.g., the biological sample is centrifuged, the biological sample is filtered, the biological sample is diluted, the biological sample is treated with reagents (e.g., digesting enzymes or reducing reagents), the biological sample is fractionated to remove more abundant proteins (e.g., proteins present at concentrations greater than 0.01 g/dL, greater than 0.02 g/dL, greater than 0.05 g/dL, greater than 0.1 g/dL, greater than 0.2 g/dL, greater than 0.5 g/dL, greater than 1.0 g/dL, greater than 2.0 g/dL, greater than 3.0 g/dL), such as, albumins, globulins (e.g., haptoglobulin, alpha2-macroglobulin, IgG, IgA, and IgM), alpha1-acid glycoprotein, apolipoprotein AI, apolipoprotein AII, complement C3, transthyretin, antitrypsin, transferrin, and fibrinogen and/or enrich for less abundant proteins, such as, any protein from Tables 1, 2, 4, and/or 5.


For example, a blood sample may be obtained, and prior to determining the level of one or more proteins, the sample may be centrifuged to remove red blood cells (i.e., to provide a plasma sample). The plasma sample may be spin filtered and diluted. Subsequently, the sample may be chromatographically separated using an immunoaffinity-based column to remove more abundant proteins (e.g., the 10-20, e.g., 10, 12, 14, 16, 18, 20 most abundant proteins) and enrich for less abundant proteins. The enriched sample may be concentrated and buffer exchanged, followed by treatment with a digesting enzyme, e.g., trypsin. Determination of protein levels may then be carried out on the processed sample.


Methods for Predicting and Monitoring Response to Kawasaki Disease Therapies

The invention further features methods for predicting response to a Kawasaki disease therapy in a subject before or after administration of one or more Kawasaki disease therapies. These methods may be carried out generally as described above or as known in the art with respect to sample collection and assay format. For example, these methods may be carried out by collecting a sample, e.g., a blood or plasma sample from a subject; measuring the level of one or more biomarkers described herein (e.g., proteins of Table 1, 2, 4, 5, 6, 7, 8, and/or 9, genes of Table 10, Table 11, Table 12, and/or Table 13, and/or glycans of Table 14, Table 15, Table 16, and/or Table 17) in the sample and/or determining the binding of IgG in the sample to a protein comprising an amino acid sequence of Table 3; comparing to a control sample; and making a prediction about whether the subject will be responsive to a Kawasaki disease therapy. The method also can be used to predict whether a subject, who has been diagnosed with Kawasaki disease, will respond positively to a Kawasaki disease therapy such as a therapeutic (e.g., IVIG) or a combination of therapeutics (e.g., IVIG and one or more anticoagulants, an anti-inflammatory agent, and/or one or more immunosuppressant drugs).


A prediction of a positive response refers to a case where the Kawasaki disease symptoms will be alleviated and/or the risk of mortality will be reduced as a result of the Kawasaki disease therapy.


In the methods of predicting response to a Kawasaki disease therapy, the level of the protein(s) binding of IgG, gene(s), and/or glycan(s) can be determined relative to a control value. A control value can be a range or average value from a normal subject or a population of normal subjects; a value from a sample from a subject or population of subjects who have undergone a Kawasaki disease therapy and have reduced symptoms following therapy; a value from the same subject before the subject was diagnosed or before the subject started treatment.


The methods of the invention can be used to predict whether a subject will be responsive to a Kawasaki disease therapy, for example, an increase in the level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 1) may indicate a positive response to a Kawasaki disease therapy. Similarly, a decrease in the level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 2) may indicate a positive response to a Kawasaki disease therapy. Also, increased binding (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) of IgG in the sample may indicate a positive response to a Kawasaki disease therapy.


Alternatively, an increase in the level (e.g., an increase by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more, or an increase by more than 1.2-fold, 1.4-fold, 1.5-fold, 1.8-fold, 2.0-fold, 3.0-fold, 3.5-fold, 4.5-fold, 5.0-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 1000-fold, or more, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 4) may indicate a poor response to a Kawasaki disease therapy. Similarly, a decrease in the level (e.g., a decrease by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, or more; or a decrease by less than 0.01-fold, 0.02-fold, 0.1-fold, 0.3-fold, 0.5-fold, 0.8-fold, or less, as compared to a reference) of the biomarker(s) (e.g., a protein of Table 5) may indicate a poor response to a Kawasaki disease therapy.


The methods of the invention can be used to predict a subject's response to a Kawasaki disease therapy and classify the subject as a “responder,” e.g., a subject with protein levels and/or binding of IgG indicative of a positive response to a Kawasaki disease therapy (e.g., IVIG), or a “non-responder,” e.g., a subject with protein levels and/or binding of IgG indicative of a poor response to a Kawasaki disease therapy (e.g., a subject that may benefit from a therapy other than, or in addition to, IVIG therapy).


The prediction can be made prior to administration of a first Kawasaki disease therapy. Alternatively, the prediction can be made after administration of the first Kawasaki disease therapy, or after administration of a first Kawasaki disease therapy but before a second Kawasaki disease therapy. Furthermore, the prediction can be made at any time during the course of a Kawasaki disease therapy.


The methods described herein can also be used to monitor Kawasaki disease status (e.g., progression or regression) during therapy or to optimize dosage of one or more therapeutic agents for a subject. For example, alterations (e.g., an increase or a decrease as compared to either the positive reference sample or the level diagnostic for Kawasaki disease) can be detected to indicate an improvement in Kawasaki disease status. In this embodiment, the levels of the protein(s), binding of IgG, gene(s), and/or glycan(s) may be measured repeatedly as a method of not only diagnosing disease, but also monitoring the treatment, prevention, or management of the disease.


In order to monitor the status of Kawasaki disease in a subject, subject samples may compared to reference samples taken early in the diagnosis of the disorder. Such monitoring may be useful, for example, in assessing the efficacy of a particular therapeutic agent (e.g., IVIG) in a subject, determining dosages, or in assessing disease progression or status. For example, levels of any of the proteins, genes, and/or glycans described herein, and/or binding of IgG, or any combination thereof can be monitored in a subject, and as the levels or activities increase or decrease, relative to control, the dosage or administration of therapeutic agents may be adjusted.


Methods of Treatment

The invention also features a method for treatment of Kawasaki disease in a subject by contacting a biological sample from the subject with one or more binding agents capable of specifically binding one or more biomarkers (e.g., one or more proteins of Table 6); determining if the level in said biological sample is changed relative to a control value; predicting a response to a Kawasaki disease therapy in said subject based on the level of one or more of said biomarkers; and, if the prediction is positive, administering a Kawasaki disease therapy.


The methods can also be used to determine the proper dosage (e.g., the therapeutically effective amount) of a therapeutic agent for the subject, the proper type of therapeutic agent, or whether a therapy should be administered.


Several therapeutic agents have been used in the treatment of Kawasaki disease. These include, without limitation, administration of IVIG (i.e., IVIG therapy); salicylates (e.g., aspirin); corticosteroids (e.g., prednisone); IL-1 receptor antagonists; anticoagulants (e.g., enoxaparin and/or clopidogrel); infliximab; or any combination thereof. Approximately 25% of subjects do not respond to IVIG treatment. These predisposed subjects are at a higher risk for developing cardiac artery aneurysms or stenosis. These subjects, as classified herein, may be administered low molecular weight heparin (LMWH), such as enoxaparin, in addition to IVIG, for an extended period of time, e.g., weeks to months. Other therapies that may be administered in addition to IVIG, include, but are not limited to, anti-TNF agents (e.g., adalimumab, infliximab, or etanercept); anti-IL treatment (e.g., anti-IL-1a, IL-1 b, IL-1 RA); statins (e.g., atorvastatin, pravastatin); corticosteroids (e.g., prednisolone, methylprednisolone); immunomodulators (e.g., cyclosporine A, methotrexate), anti-CD20 therapy (e.g., rituximab); plasma exchange, warfarin, and fibrinogen receptor glycoprotein IIb/IIIa, such as Abciximab.


Diagnostic Kits

The invention also provides test kits and devices. For example, a diagnostic test kit can include one or more binding agents and, if desired, components for detecting and/or evaluating binding between the binding agent and a biomarker. For detection, one or more of binding agents may be labeled. In further embodiments, one or more of the binding agents may be substrate-bound, such that a biomarker-antibody interaction can be established by determining the amount of label attached to the substrate following binding between the antibody and the biomarker. A conventional ELISA is a common, art-known method for detecting antibody-substrate interaction and can be provided as the kit of the invention.


A kit that determines an alteration in the level of a protein, binding of IgG, to a peptide, gene, and/or glycan relative to a reference, such as the level present in a normal control, is useful as a diagnostic kit in the methods of the invention. Such a kit or device may further include a reference sample or standard curve indicative of a positive reference or a normal control reference.


Desirably, the kit will contain instructions for the use of the kit. In one example, the kit contains instructions for the use of the kit for the diagnosis of Kawasaki disease. In yet another example, the kit contains instructions for the use of the kit to monitor therapeutic treatment or dosage regimens. In yet another example, the kit contains instructions for the use of the kit to predict outcome, response to therapy, or disease recurrence. In a further example, the instructions include one or more metrics (e.g., metrics to be used as references).


EXAMPLES

The following examples are intended to illustrate the invention. They are not meant to limit the invention in any way.


Methods

Plasma Proteomics


Proteomics on human plasma samples were carried out as follows. In order to enhance sensitivity for less abundant species, the top 14 most abundant proteins were depleted from plasma samples prior to proteomics analysis using the Agilent MARS column. Briefly, 50 μl of human plasma was spin filtered and diluted with the sample buffer and applied to a MARS column. Two fractions, eluted from the column, were collected: fraction 1 representing less abundant proteins (LA Frn1) and fraction 2 representing highly abundant serum proteins (HA Frn2). Both fractions were concentrated and buffer exchanged. LA Frn1 was digested by trypsin, followed by proteomics analysis. Lower abundant fraction-1 tryptic digests were subjected to nano-LC-MS/MS analysis. Separations were carried out using an Ultimate3000 RSLCnano system. Chromatography was carried using analytical EASY-Spray PepMap RSLC, 25 cm×75 μm id, C18, 2 μm and 100 Å nano column thermostatically controlled at 50° C. and at 300 nL/min with a linear gradient from 1% to 38% acetonitrile/water both containing 0.1% (v/v) FA for a total duration of 150 minutes. The separation step was followed by a 30 minute washing step with 99% acetonitrile/water followed by a 20 minute equilibration step with 99% water/acetonitrile both containing 0.1% (v/v) FA. 1.0 μg of each sample was injected into the column. Data dependent MS-MS was performed on the top 25 precursor ions from the full MS scan on the Orbi-Velos MS instrument.


1 μg protein from each sample was injected into the nano-LC-MS/MS system. The data quality was reproducible across all samples. Proteome discoverer with Sequest search engine was used for database searching of each sample against Uniprot human protein database. ˜450 proteins were identified from each sample. The distribution of peptide spectral matches (PSM) was plotted for each sample, and the overall assessment indicated that most samples contained equivalent numbers of PSM as observed.


Overall, ca. 440 protein signatures were acquired per sample using the set criteria.


Log ratios were calculated from the average normalized spectral counts from pairwise comparisons to get the fold change of proteins across the groups. T-test was conducted to determine the significance of the change. Further, the data were filtered to include changes with less than or equal to 0.05 T-test value.


Soluble Plasma Proteins


The concentrations of plasma IgG and IgA were determined using the Total Human IgG Immunoenzymetric Assay and the Human Immunoglobulin A Immunoenzymetric Assay from Cygnus Technologies (Southport, N.C.). The Human FcγR3B ELISA Kit from MyBioSource (San Diego, Calif.) was used to quantify the concentration of soluble CD16 in the patient samples. The concentration of α2,6-sialyltransferase (ST6GALI) in the plasma samples was quantified using the α2,6-Sialyltransferase Assay Kit from Immuno-Biological Laboratories (Minneapolis, Minn.). The plasma samples were diluted in diluents provided with each ELISA. The manufacturer's instructions supplied with the assay kits were followed for the quantitation of each analyte. Absorbance readings for all assays were determined using a SpectraMax M2 spectrophotometer (Molecular Devices, Sunnyvale, Calif.) and analysis was performed using SoftMax Pro 5.2 software (Molecular Devices).


IgG and IgA Glycomics


Methods for glycosylation analysis of Ig proteins is known in the art, for example, as described in Pucic et al, Molecular and Cellular Proteomics, 2011 October(10): 1-15.


Tryptic digests of Protein G enriched plasma fraction were subjected to nano-LC-MS/MS analysis. Two IVIg samples were used along with the KD samples as controls throughout the sample prep and data analysis. All separations were carried out using an Ultimate3000 RSLCnano system. Chromatography was carried using analytical EASY-Spray PepMap RSLC, 25 cm×75 μm id, C18, 2 μm and 100 Å nano column thermostatically controlled at 50° C. and at 300 nL/min with a linear gradient from 1% to 38% acetonitrile/water both containing 0.1% (v/v) FA for a total duration of 70 minutes. The separation step was followed by a 30 minutes washing step with 99% acetonitrile/water followed by a 20 minutes equilibration step with 99% water/acetonitrile both containing 0.1% (v/v) FA. 1.0 ug of each sample was injected on column. Data dependent MS-MS was performed on top 25 precursor ions from the full MS scan on the Orbi-Velos MS instrument.


Data analysis was performed using Xcalibur software—Qual and Quan browser. Area of each of the glycopeptides (IgG1, IgG2, IgG3/4 and IgA) was extracted from the base peak chromatogram. All the glycopeptide species m/z and retention time were captured in a processing method that was used for quantitation across all the samples. Peak integration was manually checked for accuracy in the quan browser. The raw abundances/area of all the glycopeptides for all the 30 KD samples and two IVIg controls was then exported in excel. The data was normalized based on the total glycopeptide abundance for each specific IgG (1, 2, 3/4) and IgA protein within the sample. Based on these data analysis, we reported the relative abundance of each of the IgG1, IgG2, IgG3/4, and IgA glycopeptide species per sample. Up to this point the samples were all blinded. Once we had the measurement values, then for interpretation we used the unblinded sample description wherein we could group the samples as Febrile controls, Acute phase KD, IVIg treated, and LMWH treated patient samples.


Total Plasma Glycome


To low- and high-abundant protein plasma fractions (25 μL) were added: 2 μL of sample buffer (0.625 mL of 0.5M Tris, pH 6.6, 1 mL of 10% SDS, and 3.375 mL of water), 2 μL of water, and 1 μL of 0.5M dithiothreitol (DTT), followed by incubation at 65° C. for 15 min. The samples were then alkylated by adding 1 μL of 100 mM iodoacetamide and incubated for 30 min in the dark at room temperature. The samples were then set into gel blocks by adding 22.5 μL of 30% (w/w) acrylamide/0.8% (w/v) bis-acrylamide stock solution (37.5:1.0, Protogel, National Diagnostics, Hessle, Hull, UK), 11.25 μL of 1.5M Tris (pH 8.8), 1 μL of 10% SDS, 1 μL of 10% ammonium peroxodisulfate (APS), and finally 1 μL of N,N,N,N′-tetramethyl-ethylenediamine (TEMED), mixed, and then left to set. The gel blocks were transferred to a filter plate and washed with 1 mL of acetonitrile with vortexing on a plate mixer (Sarstedt, Leicester, UK) for 10 min, followed by removal of the liquid. Washing procedure was repeated twice with 1 mL of 20 mM NaHCO3(pH 7.0) followed by 1 mL of acetonitrile. N-glycans were released by adding 50 μL of 0.1 U/mL PNGaseF (Prozyme, CA, USA) in 20 mM NaHCO3 (pH 7.0) to each sample and incubating overnight at 37° C. The released glycans were collected by washing the gel pieces with 3×200 μL of water, 200 μL of acetonitrile, 200 μL of water, and finally 200 μL of acetonitrile. The released glycans were dried, 20 μL of 1% formic acid was added, and the mixture was incubated at room temperature for 40 min and then re-dried. Samples were labeled by adding 5 μL of 2AB labeling solution, vortexed, incubated for 30 min at 65° C., vortexed again, and incubated for a further 90 min. Excess 2AB was removed using Whatman 3 MM chromatography paper cleanup: 1-cm square pieces of prewashed, dried Whatman 3 MM chromatography paper were folded into quarters and placed into a filter plate (Whatman protein precipitation plate prewashed with 200 μL of acetonitrile followed by 200 μL of water). The 5 μL of 2AB-labeled samples were applied to the paper and left to dry/bind for 15 min. The excess 2AB was washed off the paper by vortexing with 1.8 mL of acetonitrile for 15 min and then removing the acetonitrile using a vacuum manifold; this procedure was repeated four times. The labeled glycans were eluted from the paper by vortexing with 900 μL of water for 30 min and then collected by vacuum into a 2-mL 96-well plate. This was repeated with a further 900 μL of water. The eluates were dried and re-constituted in H2O/ACN (v/v 30/70). Released and labeled glycans were subsequently fractionated by normal phase chromatography. Ultra Performance Hydrophilic interaction liquid chromatography (UPLC-HILIC) was carried out on a 1.7 μm Waters BEH Glycan (150 mm×2.1 mm) column as detailed in Mittermayr et al. “Multiplexed analytical glycomics: Rapid and confident IgG N-glycan structural elucidation,” J. Prot. Res. 2011:10:3820-9 with retention times expressed as glucose units (GU) with the following conditions: Solvent A was 50 mM formic acid adjusted to pH 4.4 with ammonia solution. Solvent B was acetonitrile. The column temperature was set to 30° C. The 30 min method was used with a linear gradient of 30-47% with solvent A (=70-53% solvent B) at 0.56 mL/min for 23 min followed by 47-70% solvent A and finally reverting back to 30% solvent A to complete the run method. Samples were injected in 60% acetonitrile. Samples were run once and peaks were identified by their GU values which were compared with the GlycoBase 3.1 structural library for serum N-Glycans (http://glycobase.nibrt.ei).


Glycan HILIC data represent the relative percentage areas from HILIC profiles. Therefore, the data are compositional, and convey the relative amounts of glycan structures in a sample rather than the absolute quantities. Compositional data are subject to an awkward constant sum constraint, that is, the values sum to a constant value such as one or one hundred percent. For this reason, the logit transform was used to map the data onto real space.


Gene Expression


RNA was purified from whole blood collected in Paxgene tubes. Reverse transcription was performed on 0.5 μg of RNA with random hexamer priming (Invitrogen) and reverse transcriptase (New England Biolabs). Expression analysis of 48 genes was performed by Real Time-Quantitative Polymerase Chain Reaction (RT-qPCR) performed on the Roche LightCycler 480 II. The ΔΔCp method (Pfaff) 2001) was used to quantify transcripts using the average Cp value of housekeeping genes ACTB, GUSB and RPS14 to normalize each sample. −ΔCp was used as an arbitrary unit measure of transcript quantity.


Study Design


Subject samples selected for this study were between 2 to 33 months of age, Hispanic, males treated for Kawasaki Disease at Kawasaki Disease Center at Rady Children's Hospital.


Thirty subject samples, consisting of blood plasma, DNA, and RNA were used in the study. The Kawasaki Disease group consisted of 3 subjects that received IVIG and LOVENOX® treatment (Note: Subject 1 also received a second dose of IVIG 14 days after the first dose); and 5 subjects that received IVIG. A control group was selected consisting of 5 age and sex matched non-Kawasaki, febrile infants.


Samples were categorized according to the stage of disease or treatment into:


Group A—acute Kawasaki


Group B—sub-acute, post IVIG


Group C—on LOVENOX®;


Group D—convalescent


Febrile controls have only acute samples available.


All samples were randomized and blinded for each analytical platform that was applied.


The subject's clinical information, including treatment information, is provided in Table 21.









TABLE 21







Subject clinical information










Subject
CA











ID #
status*
Treatment**













1
2
IVIG × 2
Second IVIG given 2 weeks after initial





treatment for cardiac indications


2
4
IVIG × 2,
Second IVIG + infliximab for cardiac




infliximab
indications


3
2
IVIG × 2
Second IVIG for cardiac indications


4
2
IVIG × 2
Second IVIG for treatment resistance


5
3
IVIG × 2
Second IVIG for treatment resistance


6
2
IVIG × 2
Second IVIG for cardiac indications


7
1
IVIG × 2
Second IVIG for treatment resistance


8
3
IVIG × 2
Second IVIG for treatment resistance





*Coronary artery status:


1 = normal echo,


2 = coronary artery aneurysms,


3+ transiently dilated,


4 = giant aneurysms


**Subjects treated with second IVIG for “treatment resistance” had fever ≥ 36 h post-end of IVIG infusion;


subjects treated with second IVIG for cardiac indications had abnormal echo and therefore received 2 infusions of IVIG to maximize the anit-inflammatory benefit


The sample information is provided in Table 22.













TABLE 22







Sample information













Sample



Age



ID #
Stage
Day
Treatment
(months)

















 1A
Acute KD
4
IVIG/LMWH
2.4



 1B
Post IVIG
12





 1C
on Lovenox
21





 1D
Convalescent
693





 2A
Acute KD
8
IVIG/LMWH
3.9



 2B
Post IVIG
4





 2C
on Lovenox
13





 2D
Convalescent
NA





 3A
Acute KD

IVIG/LMWH
14.6



 3B
Post IVIG
NA





 3C
on Lovenox
37





 3D
Convalescent
364





 4A
Acute KD

IVIG
3.4



 4B
Post IVIG
6





 4D
Convalescent
26





 5A
Acute KD

IVIG
2.4



 5B
Post IVIG
16





 5D
Convalescent
33





 6A
Acute KD

IVIG
15.2



 6B
Post IVIG
12





 6D
Convalescent
21





 7A
Acute KD

IVIG
19.6



 7B
Post IVIG
21





 7D
Convalescent
490





 8A
Acute KD

IVIG
33.3



 8B
Post IVIG
15





 8D
Convalescent
22





 9A
Acute
NA
Control




10A
Acute

Control




11A
Acute

Control




12A
Acute

Control




13A
Acute

Control







High risk subjects 1-3 received IVIG/LMWH treatment.



Subjects 4-8 received IVIG treatment leading to disease resolution.



Subjects 9-13 were febrile control.






Example 1. Identification of Proteins Indicative of Kawasaki Disease

Shot-gun proteomics identified ˜450 unique proteins on average per sample. In the targeted comparison of Kawasaki disease subjects at the acute stage (1A-8A) with the febrile control (9A-13A), 39 proteins were differentially expressed with 22 down-regulated and 17 up-regulated with a p-value <0.05 as a filter. These differentially expressed proteins belong to: inflammatory response pathway (S100A9, ORM1 ↑), Statin pathway (Apolipoproteins↑), Complement/Coagulation cascades (CFH, Serpin A1, C1), and Autophagy (GSN ↑), and they were among those altered in Kawasaki compared to the Febrile control.


A list of proteins that were found to be significantly different between the Kawasaki disease group (1A-8A) and the febrile control group (9A-13A) are shown in Table 23.









TABLE 23







Proteins that are significantly different (p < 0.05) in KD group versus febrile control















Febrile
Acute






Control
KD
Log2FC


Accession #
Description
p-value
Value
Value
Acute















E7ENL6
collagen, type VI, alpha 3
0.0140
5.41
1.27
−2.1


Q5MNV8
F-box protein 47
0.0100
3.71
0.98
−1.9


P12109
collagen, type VI, alpha 1
0.0161
2.70
0.76
−1.8











P10275
androgen receptor
0.0018
4.01
1.30












Q12888
tumor protein p53 binding
0.0003
2.01
0.76
−1.4



protein 1






Q6UXB8
peptidase inhibitor 16
0.0333
10.67
4.14
−1.4


P55290
cadherin 13
0.0017
2.13
0.83
−1.4


G5E9A9
tenascin XB
0.0308
8.19
3.50
−1.2


B0UYX3
tenascin XB
0.0365
7.42
3.20
−1.2


Q07869
peroxisome proliferator-
0.0377
1.55
0.69
−1.2



activated receptor alpha






P02452
collagen, type I, alpha 1
0.0168
3.57
1.75
−1.0


P01877
Ig alpha-2 chain C region
0.0485
9.80
4.96
−1.0



(A2m marker)






P01765
Ig heavy chain V-III region TIL
0.0111
1.34
0.69
−1.0


P01876
Ig alpha-1 chain C region
0.0420
17.95
9.19
−1.0


Q6IMJ5
olfactomedin 1
0.0311
3.36
1.74
−1.0


Q5TFM2
complement factor H
0.0026
92.22
48.12
−0.9


P41222
prostaglandin D2 synthase
0.0211
2.45
1.30
−0.9



21 kDa (brain)






I3L145
sex hormone-binding globulin
0.0007
12.83
6.97
−0.9


P01593
Ig kappa chain V-I region
0.0131
3.02
1.66
−0.9


P06396
gelsolin
0.0003
70.33
39.42
−0.8


P0CG04
Ig lambda-1 chain C regions
0.0102
12.65
7.15
−0.8



(Mcg marker)






Q9NSY1
BMP2 inducible kinase
0.0130
1.34
0.76
−0.8


P01008
serpin peptidase inhibitor,
0.0250
215.36
124.42
−0.8



clade C (antithrombin),







member 1






P20742
pregnancy-zone protein
0.0430
10.65
6.20
−0.8


P19827
inter-alpha-trypsin inhibitor
0.0447
193.28
115.09
−0.7



heavy chain 1






P01617
Ig kappa chain V-II region
0.0202
1.90
1.14
−0.7


P19823
inter-alpha-trypsin inhibitor
0.0437
186.02
114.81
−0.7



heavy chain 2






P08603
complement factor H
0.0046
226.25
141.04
−0.7


Q04756
Hepatocyte growth factor
0.0203
6.81
4.42
−0.6



activator






O75882
attractin
0.0433
13.74
8.92
−0.6


P01042
kininogen 1
0.0134
97.64
63.38
−0.6


P05160
coagulation factor XIII, B
0.0401
15.43
10.20
−0.6



polypeptide






B4E1Z4
complement factor B
0.0052
213.26
149.14
−0.5


H0YFH3
complement component 1, r
0.0079
33.02
23.48
−0.5



subcomponent






P00736
complement component 1, r
0.0109
36.72
27.34
−0.4



subcomponent






Q16610
extracellular matrix protein 1
0.0263
20.87
15.70
−0.4


P09871
complement component 1, s
0.0085
40.93
31.31
−0.4



subcomponent






C9JEX1
kininogen 1
0.0195
65.08
50.06
−0.4


P13671
complement component 6
0.0163
59.56
46.05
−0.4


P02774
vitamin D binding protein
0.0259
396.66
320.83
−0.3



(group-specific component)






P01011
Alpha-1-
0.0029
387.52
536.07
0.5



antichymotrypsin (serpin







peptidase inhibitor, clade A,







member 3)






F5H5G7
lactate dehydrogenase C
0.0106
0.90
1.36
0.6


G3V3A0
Alpha-1-
0.0006
152.86
246.65
0.7



antichymotrypsin (serpin







peptidase inhibitor, clade A,







member 3)






Q8NEN0
armadillo repeat containing 2
0.0193
1.00
1.69
0.8


Q9H3U1
unc-45 homolog A (C.
0.0145
0.88
1.52
0.8



elegans)






P59665
defensin, alpha 1
0.0048
1.01
1.75
0.8


P06702
S100 calcium binding protein
0.0358
6.13
12.50
1.0



A9






P40879
solute carrier family 26 (anion
0.0467
1.13
2.38
1.1



exchanger), member 3






P43403
zeta-chain (TCR) associated
0.0094
1.01
2.15
1.1



protein kinase 70 kDa






Q9H2Y7
Zinc finger protein 106
0.0093
0.78
1.76
1.2



homolog






P04114
apolipoprotein B
0.0021
159.94
363.53
1.2


J3QLI2
Zinc finger protein 161
0.0013
1.55
3.59
1.2



homolog






O95445
apolipoprotein M
0.0095
0.79
1.84
1.2


P02655
apolipoprotein C-II
0.0446
1.66
4.20
1.3


F5GXS5
apolipoprotein F
0.0145
0.67
1.82
1.4


I3L1Y6
zinc finger protein 578
0.0402
0.67
2.14
1.7


A4UGR9
xin actin-binding repeat
0.0001
1.00
3.49
1.8



containing 2






P08519
lipoprotein, Lp (a)
0.0475
0.90
3.54
2.0


Q8N2F6
armadillo repeat containing 10
0.0069
0.56
2.54
2.2


Q8NGL3
olfactory receptor, family 5,
0.0201
1.57
7.17
2.2



subfamily D, member 14






P02763
orosomucoid 1
0.0251
17.47
162.97
3.2


P01009
Alpha-1-antitrypsin (serpin
0.0374
27.65
314.28
3.5



peptidase inhibitor, clade A,







member 1)






4843
nitric oxide synthase 2,
0.0491
0.049135
0.00028
1.8



inducible (Gene)






1088
carcinoembryonic antigen-
0.0274
0.027424
0.00684
2.2



related cell adhesion molecule







8 (Gene)






90865
interleukin 33 (Gene)
0.0432
0.043233
0.000154
3.8


3569
interleukin 6 (interferon, beta
0.0100
0.00164
0.091973
5.8



2) (Gene)






4312
matrix metallopeptidase 1
0.0115
7.84E−05
0.124912
10.6



(interstitial collagenase)







(Gene)









SERPINA3 protein is an example of a measurement that separates Febrile Controls and Acute Kawasaki Disease. SERPINA3 is considered to be an acute phase response protein observed to be increased during certain types of inflammatory response. CRP is another protein that belongs to the acute phase response group. However, CRP does not appear to consistently separate Febrile Controls from Acute Kawasaki Disease subjects. IVIG treatment appears to reduce levels of both of these proteins as evidenced by reduced levels after IVIG treatment.


Example 2. Identification of Proteins Indicative of Development of Cardiac Artery Aneurysm or Stenosis

Current standard of care for Kawasaki disease is treatment with IVIG/ASA at the time of diagnosis at the acute stage. As there is no test or diagnostic tool to identify subjects “at risk” for developing cardiac aneurysm at the acute stage, the aneurysm treatment (LOVENOX®) is generally delayed until echocardiogram imaging is provided later in the course of disease, often when a coronary artery aneurysm has already occurred. A test or diagnostic tool that identifies subjects “at risk” earlier could prevent the development of coronary artery aneurysms and the resulting long term effects (e.g., increased risk of heart attack and other cardiovascular events later in life).


To identify proteins indicative of development of cardiac artery aneurysms or stenosis, a group of Kawasaki disease subjects (1A, 2A, 3A) who were subjected to IVIG/LOVENOX® treatment (based on the clinical outcome) was compared with a group of subjects that were treated with IVIG only (4A, 5A, 6A, 7A, 8A). All subjects in the IVIG/LOVENOX® group developed coronary aneurysm. These subjects may be genetically predisposed to developing coronary artery aneurysm even with the appropriate standard of care treatment.


Ten proteins were significantly altered in the high risk group: three were up-regulated (PROC, F11, APOF) and seven down-regulated (CD44, ANKRD26, LAMP2, BCAM, MMRN1, TGFBI, TET2). The differential levels of these proteins in subjects that developed coronary artery aneurysms or stenosis indicates these proteins are useful for selecting or classifying subjects predisposed to coronary artery aneurysms or stenosis. This early selection may allow for treatment with anticoagulants (e.g., enoxaparin and/or clopidogrel) or other therapies (e.g., infliximab, cyclosporine, and/or prednisone) to begin before the development of coronary artery aneurysms or stenosis, rather than in response to their development as is the current practice.


These protein identifiers were mapped to the following functions:

    • a. Proteins related to autophagy—CD44 and LAMP2
    • b. Proteins from complement/coagulation cascades—PROC and F11 are lower in IVIG/LOVENOX® group versus IVIG groups, suggesting that these protein markers for wound healing are present in higher expression in the IVIG responders.
    • c. Cell adhesion related proteins such as BCAM, TGFB1, and MMRN1 show higher expression in the high risk group as compared with the IVIG responders.


A list of proteins that were found to be significantly different between the IVIG/Lovenox® group (1A-3A) and the IVIG only group (4A-8A) are shown in Table 19.









TABLE 24







Proteins that are significantly different (p < 0.05) in IVIG/


LOVENOX ® group versus IVIG only group














IVIG/

Log



Accession

LOVENOX ®
IVIG
fold
p-


No
Description
Value
value
change
value















P00747
Plasminogen
43.78
60.98
−0.5
0.0408


P04070
Vitamin K-dependent protein C
0.99
2.81
−1.5
0.0014


P03951
Coagulation factor XI
2.17
5.25
−1.3
0.0235


F5GXS5
Apolipoprotein F
0.99
2.32
−1.2
0.0326


F5H5G7
L-lactate dehydrogenase
1.60
1.22
0.4
0.0064


E7EPC6
CD44 antigen
3.61
2.08
0.8
0.0148


Q9UPS8
Ankyrin repeat domain-containing
5.23
2.56
1.0
0.0199



protein 26






B4E2S7
Lysosome-associated membrane
2.01
0.98
1.0
0.0234



glycoprotein 2






P50895
Basal cell adhesion molecule
2.39
1.10
1.1
0.0018


Q13201
Multimerin-1
2.84
1.23
1.2
0.0035


G8JLA8
Transforming growth factor-beta-
7.12
2.94
1.3
0.0206



induced protein ig-h3






E7EQS8
Methylcytosine dioxygenase TET2
2.88
0.98
1.6
0.0067


P01011
Alpha-1-antichymotrypsin
610.74
491.26
0.3
0.0110


P01034
Cystatin-C
2.62
1.96
0.4
0.0197









In an effort to identify further proteins indicative of development of cardiac artery aneurysms or stenosis, a group of Kawasaki disease subjects (1, 2, 3, 4, and 6) who developed aneurysms based on electrocardiogram imaging was compared with a group of subjects that either did not develop aneurysms or were only dilated and not considered to have developed full aneurysms (5, 7, and 8). The subjects that developed aneurysms may be genetically predisposed to developing coronary artery aneurysm even with the appropriate standard of care treatment.


Student's t-test were used to determine if any significant differences were observed between aneurysm and non-aneurysm groups. Of the 647 measurements, 10 and proteins had a p-value less than 0.05 (see Table 25).









TABLE 25







Proteins that are significantly different (p < 0.05) in


aneurysm group versus non-aneurysm group













Accession

Log2 FC


Gene Symbol
Analytic
Number
P-value
no aneurysm/aneurysm














HGFAC
PRO
Q04756
1.15E−02
−0.6


BST1
PRO
Q10588
4.23E−02
−1


AFTPH
PRO
Q6ULP2
4.86E−02
2


DTHD1
PRO
Q6ZMT9
4.35E−02
0.9


MASP2
PRO
O00187
4.27E−02
−0.6


XIRP2
PRO
A4UGR9
4.35E−02
0.6


MTA3
PRO
E7EV10
6.65E−05
1.7


hCG_2014417
PRO
B7Z718
3.45E−02
−1.4


TET2
PRO
E7EQS8
3.21E−02
1.4


TIAM1
PRO
F5GZ53
3.95E−02
−1.6









Example 3. Identification of Proteins with Different Binding to Plasma IgG Between Samples of Acute Kawasaki Disease and Febrile Control

A 10K random peptide array was tested for IgG reactivities in plasma samples of KD. A total of 68 out of 10,000 peptides were found to have significantly different binding to plasma IgG between acute KD and febrile control (p<0.05).


Using the 68 peptides, an agglomerative hierarchical clustering technique (via Ward's minimum variance method) was used to naturally break the data into a hierarchy of “similar” clusters. Here, the natural break created two clusters. The 68 peptides identified have the sequences of SEQ ID NOs: 1-68, provided in Table 3, supra. The 68 peptides were attached to the microarray by cysteine-serine-glycine linkers. The peptides (including the linker sequence) are listed in Table 26.









TABLE 26







Peptides identified with different IgG


binding between febrile control and


Kawasaki disease acute stage samples









SEQ ID NO:
Peptide Sequence
p-value





 69
CSGAKFLGQSTYIAGYHQVD
7.67E−07





 70
CSGFWSKMKPSEEYTTFYRD
0.001





 71
CSGFDRSDYMSFHLDDNITI
0.001





 72
CSGIRIETPYYKDTEDGKYF
0.001





 73
CSGLGLLQAITRNSWVDSAF
0.001





 74
CSGKHWEFMQFDIGYIYEKF
0.001





 75
CSGNIPSNQHATEIQVDGYH
0.002





 76
CSGENFEYHLYDSMIGYEVH
0.002





 77
CSGERPDPATYFMPGRDDQY
0.002





 78
CSGDNAPYYYREEWHKEFNK
0.003





 79
CSGYHWDVQNTFYSMLMLPS
0.003





 80
CSGGDFSDYAPTLTQKASYG
0.003





 81
CSGQYAFHNLDQNGTVFGNR
0.003





 82
CSGYVTNMMINMNYSSLSYS
0.003





 83
CSGHWVLSDGYREVYSYNSY
0.003





 84
CSGAYHSQLYIDYKDTEWFY
0.004





 85
CSGDHPYFVIWDRYKPVHTY
0.004





 86
CSGHEHPPYLGMTAYELAQD
0.004





 87
CSGGQWSGQGYWYDPFDNMK
0.004





 88
CSGTTHFLKDRFESTNHDVY
0.004





 89
CSGVEDPRVGHSLFQDANYY
0.004





 90
CSGDPVQIFNTAEHSGPYIR
0.004





 91
CSGHDHFRGGKFILSTQAIW
0.005





 92
CSGNYPLPKYYYNWFEPRVW
0.005





 93
CSGQYDDPDWQIHYKLEARG
0.005





 94
CSGQDPYMDLHYDKNQIEQA
0.006





 95
CSGRGPHNFEIAETDAQMIE
0.006





 96
CSGDAYTNQDISEEEHMHRY
0.006





 97
CSGGALEWIYYAGPKPGYWE
0.006





 98
CSGPHSTHQIFYKSYETDMA
0.007





 99
CSGIYWGPMSTGHLPSQAQF
0.007





100
CSGYTHFWWLDKYMRYEVAT
0.007





101
CSGFLTEYYEYQNNLFHAFR
0.007





102
CSGSRGDAAAWGILFDANWK
0.008





103
CSGAMLGGMWAAYYPFPVPG
0.008





104
CSGYYWATGPEGPFRHPGAR
0.008





105
CSGDMEFTVFDIDMEKHYKY
0.008





106
CSGHYWDYQQLGFQGHLDHR
0.009





107
CSGDQHFVWGPTGRAPMNYG
0.009





108
CSGTNGFHIPFYSDFQSAAA
0.009





109
CSGMKYDVWKFYNGDDMRVS
0.009





110
CSGWYDNMYKTGFYRMYLLT
0.009





111
CSGEFQDYNHSDVNMSNHPY
0.009





112
CSGYIPEQQPHEADNLYKDA
0.010





113
CSGFYTLPHRPLYYYGYVAS
0.010





114
CSGHRWLEEANTEYMTMNSI
0.010





115
CSGGPINAYQKQDYSIEPEH
0.010





116
CSGAGDQYVRIDKSTRISNI
0.011





117
CSGSMKTVEADWYTYEPWWH
0.011





118
CSGNHIYIVQTAYGVTGETS
0.011





119
CSGMFAYHRAWPVWSSVLHV
0.012





120
CSGWPNPYFYHKKDTYWAHY
0.012





121
CSGNYDHLLGQYPIRNWWSL
0.012





122
CSGESMDVVWPYGYKFTQYW
0.012





123
CSGTHMEHDFHIPLEMYKYM
0.012





124
CSGVTPLEIIEQIREHLDIK
0.013





125
CSGLQKPFDYYMKEWQVDNE
0.013





126
CSGVMQRWPENHFLQTHYDD
0.013





127
CSGWWNWRSAYKEGDVAYPS
0.013





128
CSGGWVLDDWSSHHINYYIE
0.013





129
CSGFSPHYQYVGLFPYVKYI
0.013





130
CSGANLVYMWGSAVHTSDPQ
0.013





131
CSGSPVMYSIANYKYQTMHL
0.013





132
CSGDQEISYLNSHTDLFVGR
0.014





133
CSGYEPTDVYLTYRKLATKD
0.014





134
CSGPVPQERTDLFTGAHRAL
0.014





135
CSGREVYHEIKSGRAIEIYM
0.014





136
CSGHDAIWYDWNPYPSKHES
0.014









Example 4. Identification of Biomarkers Indicative of Development of Cardiac Artery Aneurysm or Stenosis

The predisposition for development of aneurysms can also be examined by looking at the difference in gene expression between the aneurysm group (Subjects 1 and 2) and the non-aneurysm group (Subjects 3-8). Measurements that show persistently different values in the Sub-acute samples in Subjects 1 and 2 were identified by selecting those measurements where the absolute value of the standard normal deviate was greater than 2.58 for both subjects. (Z=2.58 is the critical value for the two-tailed 99% confidence interval of the normal distribution). Sixty three measurements were observed to be persistently perturbed, either up or down, in subjects 1 and 2 at the sub-acute phase as shown in Table 27.









TABLE 27







Biomarkers that are differentially expressed in aneursym


group versus non-aneurysm group after treatment with IVIG






















Sub-
Sub-
Sub-







Acute
Acute
Acute
acute
acute
Acute
Conv.
Conv.
Conv.




Subj.
Subj.
Subj.
Subj.
Subj.
Subj.
Subj.
Subj.
Subj.


Measurement
Anal.
1
2
3-8
1
2
3-8
1
2
3-8




















ACTB
PRO
3.46
5.74
2.67
2.67
3.84
0.82
5.74
2.80
0.0


AFM
PRO
−3.35
−3.47
4.03
−3.41
−3.22
0.70
−2.42
−0.46
0.0


AHSG
PRO
−2.86
−2.63
2.63
−3.27
−4.57
1.42
−4.96
−1.66
0.0


AR
PRO
10.28
−3.67
−1.38
−9.48
−5.43
−2.42
0.30
−2.31
0.0


BCHE
PRO
−0.30
−4.10
−1.58
−4.66
−4.19
−0.36
−3.38
−1.89
0.0


BTD
PRO
−3.87
−1.67
1.99
−3.10
−3.19
2.02
−2.04
0.62
0.0


C9
PRO
4.49
3.84
4.15
4.18
3.04
1.31
3.30
1.97
0.0


CD80
GE
−5.03
−5.08
−1.53
−4.33
−5.78
−0.98
−0.38
NA
0.0


CDH13
PRO
−2.02
−4.53
9.51
−4.41
−4.89
−0.98
−1.82
0.78
0.0


CEACAM8
GE
4.98
8.87
−1.69
3.80
3.60
−0.45
4.51
NA
0.0


CHL1
PRO
−1.14
−1.43
−0.24
−2.82
−2.78
0.36
−2.05
−2.11
0.0


CRP
PRO
4.65
4.17
2.89
3.30
3.08
−0.62
2.24
1.73
0.0


CTSD
PRO
12.48
12.56
−0.22
11.18
10.87
2.05
−0.18
11.43
0.0


F10
PRO
1.09
1.23
4.05
−3.20
−3.35
0.72
−1.31
0.51
0.0


F9
PRO
2.74
0.54
3.42
−4.92
−6.45
1.32
−0.99
2.45
0.0


GP12LA
GLY
−4.68
−5.39
−5.13
−3.96
−8.08
0.52
−3.08
NA
0.0


GP13HA
GLY
11.39
11.40
−6.67
−7.83
−6.26
2.76
−5.28
NA
0.0


GP14HA
GLY
−0.93
−1.81
−2.63
−2.86
−3.30
1.99
−1.10
NA
0.0


GP14LA
GLY
−4.56
−5.34
−2.08
−2.78
−3.90
0.23
−1.44
NA
0.0


GP16LA
GLY
−4.83
−5.61
−3.35
−9.05
−5.29
0.14
−3.37
NA
0.0


GP18HA
GLY
−9.30
11.24
−6.92
−5.11
−5.57
−5.29
−4.19
NA
0.0


GP18LA
GLY
−1.99
−9.33
−3.58
−5.33
−9.96
−0.07
−2.99
NA
0.0


GP19LA
GLY
−1.38
−4.51
−2.25
−2.92
−5.86
−0.21
−0.62
NA
0.0


GP20HA
GLY
−6.37
−3.41
−7.20
16.34
10.72
−2.93
−7.11
NA
0.0


GP20LA
GLY
−9.24
−7.52
−0.89
16.51
11.69
−1.55
−5.65
NA
0.0


GP24LA
GLY
−2.96
−2.91
−5.57
−5.51
−3.13
−0.81
−0.75
NA
0.0


GP29LA
GLY
−2.36
1.15
−5.36
−6.00
3.84
−1.62
−0.95
NA
0.0


GP32HA
GLY
−4.04
−3.19
−1.63
−6.21
−5.19
−0.38
−0.34
NA
0.0


GP32LA
GLY
−3.04
−4.29
−1.70
−4.11
−4.48
0.29
−1.29
NA
0.0


GP34HA
GLY
−2.44
−2.13
2.25
−4.35
−3.66
−2.22
0.46
NA
0.0


GP34LA
GLY
−2.58
−3.32
−3.65
−3.32
−3.58
−0.18
−0.45
NA
0.0


GP37HA
GLY
2.57
3.33
−1.24
2.72
3.23
−0.59
2.47
NA
0.0


GP38LA
GLY
−1.61
−2.65
−5.11
−4.93
3.33
−0.91
0.78
NA
0.0


GP3LA
GLY
−5.97
−3.02
−4.26
−3.01
−4.92
0.74
−4.69
NA
0.0


GP42HA
GLY
3.26
4.01
−1.60
2.69
3.69
−0.15
2.67
NA
0.0


GP43HA
GLY
5.61
6.14
0.12
5.85
6.18
−0.34
3.13
NA
0.0


GP43LA
GLY
3.74
4.22
−5.21
3.90
4.48
−0.99
2.11
NA
0.0


GP44HA
GLY
7.89
9.42
−0.60
8.49
9.84
−0.21
3.32
NA
0.0


GP44LA
GLY
3.62
5.22
−7.90
3.83
5.58
5.31
1.38
NA
0.0


GP9HA
GLY
4.83
5.35
−4.02
3.99
5.20
0.85
−2.26
NA
0.0


GP9LA
GLY
2.69
6.69
−9.94
4.12
6.16
−0.37
−2.38
NA
0.0


GSN
PRO
−4.66
−6.50
4.79
−4.15
−4.87
2.93
−2.77
−0.87
0.0


GUSB
GE
−4.91
−4.67
−4.48
−3.96
−5.12
−0.39
−2.03
NA
0.0


HP
PRO
1.45
2.93
2.93
2.78
2.79
1.40
2.57
1.21
0.0


HPR
PRO
1.91
3.90
2.36
3.79
3.72
1.23
3.46
0.97
0.0


IgA
ELISA
0.26
10.99
−2.98
3.88
11.39
−1.47
7.17
NA
0.0


LRG1
PRO
6.05
6.39
3.40
3.37
3.93
0.93
2.43
3.62
0.0


LUM
PRO
−1.79
−4.21
4.43
−6.10
−6.03
1.65
−5.21
−0.78
0.0


MMP9
PRO
8.18
5.57
−7.51
2.96
3.82
−0.42
3.29
NA
0.0


NID1
PRO
0.01
4.30
4.08
2.66
4.15
−0.53
0.16
−1.98
0.0


ORM1
PRO
13.42
16.46
−0.61
9.34
13.34
0.21
1.68
−1.78
0.0


ORM2
PRO
5.79
13.52
2.75
6.30
6.85
2.31
4.60
0.23
0.0


PROC
PRO
−3.03
−4.13
2.73
−4.21
−2.81
−0.36
−1.64
−1.00
0.0


S100A12
GE
9.08
6.74
−1.11
3.91
5.55
−1.57
4.74
NA
0.0


S100A8
PRO
3.78
4.69
3.36
3.21
3.88
0.85
3.93
2.67
0.0


SERPINA4
PRO
−6.97
−4.28
1.85
−5.80
−3.28
0.48
−2.53
−0.15
0.0


ST6GAL1
ELISA
2.25
5.80
−3.01
3.70
6.25
−1.47
4.86
NA
0.0


TNXB
PRO
0.65
−4.38
3.15
−3.77
−3.47
0.07
−6.52
−0.27
0.0


TNXB_1
PRO
0.37
−5.16
0.45
−3.54
−3.90
0.83
−7.82
−0.62
0.0


TTN
PRO
−0.04
0.53
0.51
12.01
−2.65
1.11
0.26
−1.27
0.0


VCAN
GE
2.75
3.57
−1.37
4.53
3.68
−0.32
4.50
NA
0.0










PRO, protein from shot-gun proteomics; ELISA, soluble plasma protein; GE, gene expression; GLY, serum protein glycans; GP, fraction number; HA, high-abundant fraction; LA, low abundant fraction.


Example 5. Identification of Biomarkers Indicative of Response to IVIG Treatment

Measurements made on samples from KD subjects were analyzed to examine the effect of IVIG treatment. Acute phase samples were compared to samples collected after IVIG treatment.


A total of 690 analytes per sample from 21 biological samples with the complete data sets was used for the analysis. Samples were divided into an “Acute Disease” group and a “Sub-acute” group. Differences between the two groups were examined by Student's t-tests after variance stabilizing transformation. Transformations were performed by analytic: plasma proteomics, log2; RT-qPCR −ΔCp, none; plasma protein ELISA, none; plasma glycans, log2; Ig glycopeptides, logit. A total of 157 measurements with P≤0.01 were considered to be significantly different between the two groups (Acute Value vs. Sub-acute Value) and are listed in table 28.









TABLE 28







Biomarkers that are differentially expressed in acute stage versus after treatment with IVIG















Accession








No/


Acute
Post-




glycan


Stage
treatment


Analyte
Analytic
structure
Gene name
p-value
Value
Value
















CRP
PRO
P02741
C-reactive protein,
1.07E−09
15.7
0.9





pentraxin-related


HPR
PRO
P00739
haptoglobin-related
6.21E−09
107.2
9.02





protein


ITIH2
PRO
P19823
inter-alpha-trypsin
3.22E−08
122.08
259.05





inhibitor heavy chain 2


HP
PRO
P00738
haptoglobin
3.85E−08
249.76
29.02


GSN
PRO
P06396
gelsolin
5.31E−08
41.19
73.46


AFM
PRO
P43652
afamin
5.46E−08
30.22
74.24


AMBP
PRO
P02760
alpha-1-
2.27E−07
31.07
59.83





microglobulin/bikunin





precursor


FCGR1A
GE
2209
Fc fragment of IgG,
2.90E−07
−0.74
−3.72





high affinity Ia, receptor





(CD64)


BTD
PRO
F8W1Q3
biotinidase
4.26E−07
4.82
8.22


SERPINA4
PRO
P29622
serpin peptidase
6.12E−07
5.84
14.94





inhibitor, clade A





(alpha-1 antiproteinase,





antitrypsin), member 4


C9
PRO
P02748
complement
6.66E−07
50.32
32.85





component 9


ORM2
PRO
P19652
orosomucoid 2
6.85E−07
31.78
4.93


S100A12
GE
6283
S100 calcium binding
1.42E−06
0.76
−2.41





protein A12


LUM
PRO
P51884
lumican
1.51E−06
15.28
26.94


CDH13
PRO
P55290
cadherin 13
2.31E−06
0.8
1.96


ORM1
PRO
P02763
orosomucoid 1
3.15E−06
159.54
8.7


MMP9
GE
4318
matrix metallopeptidase
3.36E−06
4.31
2.17





9


GP13HA
GLY
A2[3]BG1S[3]1

3.77E−06
1.29
1.95




A2[3]BG1S[6]1




M7 D1




FA2[3]G1S[3]1


CHL1
PRO
O00533
cell adhesion molecule
4.59E−06
0.93
4.93





L1 -like


AR
PRO
P10275
androgen receptor
6.17E−06
1.14
3.14


KNG1
PRO
P01042
kininogen 1
6.94E−06
67.17
115.34


LRG1
PRO
P02750
leucine-rich alpha-2-
7.32E−06
49.06
21.97





glycoprotein 1


HGFAC
PRO
Q04756
HGF activator
1.62E−05
4.17
9.41


S100A9
PRO
P06702
S100 calcium binding
2.52E−05
13.5
2.98





protein A9


ITIH1
PRO
P19827
inter-alpha-trypsin
2.94E−05
127.79
248.86





inhibitor heavy chain 1


MST1
PRO
P26927
macrophage stimulating
3.10E−05
11.58
18.42





1 (hepatocyte growth





factor-like)


IGFALS
PRO
P35858
insulin-like growth
3.49E−05
9.52
20.4





factor binding protein,





acid labile subunit


SERPINA3
PRO
G3V3A0
serpin peptidase
3.53E−05
227.39
124.78





inhibitor, clade A





(alpha-1 antiproteinase,





antitrypsin), member 3


PGLYRP2
PRO
Q96PD5
peptidoglycan
3.69E−05
28.53
47.95





recognition protein 2


GP18HA
GLY
FA2G2S[6]1

3.86E−05
4.04
6.66


GP12LA
GLY
FA2BG2

4.25E−05
0.06
0.09


TNFRSF1A
GE
7132
tumor necrosis factor
4.69E−05
−3.89
−5.19





receptor superfamily,





member 1A


TNXB
PRO
G5E9A9
tenascin XB
5.35E−05
3.02
9.09


CLU
PRO
P10909
clusterin
5.84E−05
22.8
32.62


AHSG
PRO
P02765
alpha-2-HS-
6.16E−05
195.41
284.02





glycoprotein


SERPINA3
PRO
P01011
serpin peptidase
6.35E−05
486.42
298.31





inhibitor, clade A





(alpha-1 antiproteinase,





antitrypsin), member 3


LBP
PRO
P18428
lipopolysaccharide
6.53E−05
10.3
4.44





binding protein


GPLD1
PRO
P80108
glycosylphosphatidylino
6.79E−05
2.89
8.13





sitol specific





phospholipase D1


TNXB
PRO
B0UYX3
tenascin XB
6.79E−05
2.69
8.64


RBP4
PRO
Q5VY30
retinol binding protein
7.66E−05
20.88
70.17





4, plasma


FBLN1
PRO
B1AHL2
fibulin 1
7.67E−05
10.65
16.87


GP20LA
GLY
A2G2S[3, 6]2

7.96E−05
4.45
6.27


ST6GAL1
ELISA
NA
ST6 beta-
8.04E−05
33.66
11.47





galactosamide alpha-





2,6-sialyltranferase 1


GP18LA
GLY
FA2G2S[6]1

8.97E−05
1.25
1.58


VCAN
GE
1462
versican
1.02E−04
−0.85
−2.62


SERPINC1
PRO
P01008
serpin peptidase
1.15E−04
118.4
195.9





inhibitor, clade C





(antithrombin), member





1


GP12HA
GLY
FA2BG2

1.32E−04
0.29
0.54


HBA1
PRO
P69905
hemoglobin, alpha 1
1.35E−04
88.8
4.91


BCHE
PRO
P06276
butyrylcholinesterase
1.42E−04
4.48
9.23


S100A8
PRO
P05109
S100 calcium binding
1.52E−04
4.57
2.22





protein A8


HSPA5
PRO
P11021
heat shock 70kDa
1.75E−04
1
2.05





protein 5 (glucose-





regulated protein,





78kDa)


KLKB1
PRO
H0YAC1
kallikrein B, plasma
 2.0E−04
13.66
21.26





(Fletcher factor) 1


GP41LA
GLY
A4G4S[3, 3, 3, 3]4

2.11E−04
0.63
0.35


CSF2RB
GE
1439
colony stimulating
2.15E−04
−2.41
−4.16





factor 2 receptor, beta,





low-affinity





(granulocyte-





macrophage)


HBD
PRO
P02042
hemoglobin, delta
2.45E−04
40.53
3.68


CFH
PRO
P08603
complement factor H
2.55E−04
143.84
215.03


GP11HA
GLY
FA2G2

2.61E−04
2.84
4.8


GP19HA
GLY
FA2BG2S[3]1;

3.24E−04
0.76
1.44




FA2BG2S[6]1


GUSB
GE
2990
glucuronidase, beta
3.46E−04
−5.18
−4.9


SERPINA1
PRO
P01009
serpin peptidase
3.55E−04
264.17
21.64





inhibitor, clade A





(alpha-1 antiproteinase,





antitrypsin), member 1


FCGR3A
ELISA
NA
Fc fragment of IgG, low
3.74E−04
8.95
5.12





affinity IIIa, receptor





(CD16a)


CFH
PRO
Q5TFM2
complement factor H
4.03E−04
50.22
93.38


GP42LA
GLY
A4F1G4S[3, 3, 3, 6]4

 4.1E−04
1.2
0.5


GP42HA
GLY
A4F1G4S[3, 3, 3, 6]4

4.16E−04
1.78
0.63


GP3HA
GLY
M5;

4.22E−04
1.26
1.99




FA2B;




A2[6]G1


CEP70
PRO
Q8NHQ1
centrosomal protein
4.45E−04
1.93
3.55





70kDa


GP10HA
GLY
A2BG2

4.49E−04
0.03
0.06


HBG2
PRO
P69892
hemoglobin, gamma G
4.61E−04
18.05
1.45


HBG1
PRO
P69891
hemoglobin, gamma A
4.63E−04
18.05
1.45


GP41HA
GLY
A4G4S[3, 3, 3, 3]4

4.75E−04
0.9
0.47


HBB
PRO
P68871
hemoglobin, beta
5.42E−04
81.49
7.7


IL1B
GE
3553
interleukin 1, beta
5.71E−04
−3.25
−5.13


GP43LA
GLY
A4G4LacS[3, 3, 3, 6]4

5.71E−04
1.27
0.35


IL1RN
GE
3557
interleukin 1 receptor
6.60E−04
−1.59
−3.34





antagonist


F13B
PRO
P05160
coagulation factor XIII,
6.85E−04
10.83
20.03





B polypeptide


GP5HA
GLY
FA2[6]G1

7.79E−04
2.68
4.93


S100A8
GE
6279
S100 calcium binding
7.99E−04
1.03
−1.14





protein A8


APOH
PRO
P02749
apolipoprotein H (beta-
8.13E−04
87.26
130.48





2-glycoprotein I)


FBLN1
PRO
P23142
fibulin 1
8.21E−04
10.7
16.65


GP43HA
GLY
A4G4LacS[3, 3, 3, 6]4

8.23E−04
1.28
0.32


THBS4
PRO
P35443
thrombospondin 4
8.79E−04
3.84
8.92


GP37HA
GLY
A4F1G3S[3, 3, 3]3

9.41E−04
0.48
0.25




A4F1G3S[3, 3, 6]3




A4F1G3S[3, 6, 6]3


FN1
PRO
F8W7G7
fibronectin 1
9.44E−04
42.92
111.05


GP44HA
GLY
A4F3G4S[3, 3, 3, 3]4

9.51E−04
0.9
0.24


IGF2
PRO
P01344
insulin-like growth
1.01E−03
0.87
1.77





factor 2 (somatomedin





A)


S100A11
GE
6282
S100 calcium binding
1.03E−03
2.62
1.19





protein A11


FN1
PRO
P02751
fibronectin 1
1.04E−03
47.41
120.85


PROC
PRO
P04070
protein C (inactivator of
1.06E− 
2.07
3.68





coagulation factors Va
03





and Villa)


GP44LA
GLY
A4F3G4S[3, 3, 3, 3]4

1.15E−03
0.47
0.11


GP6HA
GLY
FA2[3]G1

1.32E−03
0.95
1.94


GP13LA
GLY
A2[3]BG1S

1.41E−03
0.43
0.66




[3]1; A2[3]BG1S[6]1;




M7 D1;




FA2[3]G1S[3]1


KNG1
PRO
C9JEX1
kininogen 1
1.56E−03
51.86
65.54


GP24LA
GLY
A2BG2S[6, 6]2

1.63E−03
1.66
2.03


SEPP1
PRO
P49908
selenoprotein P,
1.76E−03
2.97
4.76





plasma, 1


GP8HA
GLY
FA2[3]BG1;

1.85E−03
0.81
1.12




M6 D3;


NRP1
PRO
O14786
neuropilin 1
1.91E−03
1.26
2.36


NCAM1
PRO
E9PLH7
neural cell adhesion
1.94E−03
2.16
3.79





molecule 1


PKDREJ
PRO
Q9NTG1
polycystin (PKD) family
1.97E−03
2.37
0.9





receptor for egg jelly


TNFRSF1B
GE
7133
tumor necrosis factor
1.99E−03
−1.94
−2.88





receptor superfamily,





member 1B


KIF20B
PRO
Q96Q89
kinesin family member
2.04E−03
4.47
2.22





20B


LYZ
PRO
P61626
lysozyme
2.05E−03
1.66
0.78


COL1A1
PRO
P02452
collagen, type I, alpha 1
2.05E−03
1.8
3.88


GP7HA
GLY
FA2[6]BG1

2.12E−03
0.69
1.22


GP23HA
GLY
A2G2S[3, 6]2

 2.4E−03
38.79
32.23


A1BG
PRO
P04217
alpha-1-B glycoprotein
2.47E−03
100.11
131.28


F13A1
PRO
P00488
coagulation factor XIII,
2.52E−03
6.73
14.07





A1 polypeptide


ACTC1
PRO
P68032
actin, alpha, cardiac
 2.6E−03
5.8
3.25





muscle 1


ECM1
PRO
Q16610
extracellular matrix
2.62E−03
14.69
19.83





protein 1


GP32LA
GLY
A3G3S[3, 3, 6]3

2.63E−03
0.65
1.14


TNFSF13B
GE
10673 
tumor necrosis factor
2.64E−03
−1.73
−2.71





(ligand) superfamily,





member 13b


APOC1
PRO
P02654
apolipoprotein C-I
2.74E−03
1.65
3.27


GP35HA
GLY
A3F1G3S[3, 3, 3]3

2.79E−03
2.87
1.96


CPB2
PRO
Q96IY4
carboxypeptidase B2
2.79E−03
4.11
7.78





(plasma)


ATRN
PRO
O75882
attractin
2.85E−03
7.93
12.91


GP26HA
GLY
FA2BG2S[3, 6]2

2.86E−03
0.65
0.99




FA2BG2S[6, 6]2


ZNF217
PRO
O75362
zinc finger protein 217
2.89E−03
3.95
7.72


SHBG
PRO
I3L145
sex hormone-binding
2.93E−03
8
13.57





globulin


PZP
PRO
P20742
pregnancy-zone protein
3.05E−03
7.04
16.74


ITIH3
PRO
Q06033
inter-alpha-trypsin
3.20E−03
98.25
59.4





inhibitor heavy chain 3


COL11A2
PRO
H0YHY3
collagen, type XI, alpha
3.37E−03
0.93
2.24





2


SELL
PRO
P14151
selectin L
3.41E−03
4.98
7.56


ENPP2
PRO
Q13822
ectonucleotide
3.59E−03
1.5
3.99





pyrophosphatase/





phosphodiesterase 2


GP5LA
GLY
FA2[6]G1

3.62E−03
0.06
0.08


GP19LA
GLY
FA2BG2S[3]1

3.73E−03
0.17
0.21




FA2BG2S[6]1


CD80
GE
 941
CD80 (costimulatory
3.78E−03
−9.17
−8.24





molecule)


F2
PRO
P00734
coagulation factor II
3.80E−03
88.77
118.4





(thrombin)


ITIH4
PRO
Q14624
inter-alpha-trypsin
3.99E−03
396.21
362.86





inhibitor heavy chain





family, member 4


ACTBL2
PRO
Q562R1
actin, beta-like 2
4.08E−03
3.78
2.58


ITIH4
PRO
B7ZKJ8
inter-alpha-trypsin
4.08E−03
396.42
362.46





inhibitor heavy chain





family, member 4


KLRG1
GE
10219 
killer cell lectin-like
4.11E−03
−3.67
−2.9





receptor subfamily G,





member 1


ITIH4
PRO
E9PGN5
inter-alpha-trypsin
4.17E−03
395.42
362.13





inhibitor heavy chain





family, member 4


HBE1
PRO
P02100
hemoglobin, epsilon 1
4.51E−03
11.98
1.25


IgA
ELISA
NA
IgA
4.58E−03
1.11
0.41


GP6LA
GLY
FA2[3]G1

4.79E−03
0.01
0.03


GP2HA
GLY
FA2

4.85E−03
2.98
4.99


APOD
PRO
P05090
apolipoprotein D
5.18E−03
0.94
1.89


CRY2
PRO
Q49AN0
cryptochrome 2
5.32E−03
2.37
1.18


PLG
PRO
P00747
plasminogen
5.42E−03
57.53
72.3


PPP6R2
PRO
B3KMJ7
protein phosphatase 6,
5.45E−03
4.28
0.65





regulatory subunit 2


DOCK3
PRO
Q8IZD9
dedicator of cytokinesis
5.47E−03
2.6
0.84





3


FCGR2A
GE
2212
Fc fragment of IgG, low
5.73E−03
2.06
0.48





affinity Ila, receptor





(CD32)


CCR2
GE
729230 
chemokine (C-C motif)
6.66E−03
−2.4
−3.4





receptor 2


MASP1
PRO
P48740
mannan-binding lectin
6.69E−03
7.73
12.25





serine peptidase 1





(C4/C2 activating





component of Ra-





reactive factor)


GP31HA
GLY
A3G3S[3, 3, 3]3

6.74E−03
0.42
0.26


ACTB
GE
 60
actin, beta
7.45E−03
4.74
4.15


CTSD
PRO
P07339
cathepsin D
7.61E−03
4.15
0.83


TNF
GE
7124
tumor necrosis factor
7.65E−03
−4.82
−5.53


CEP290
PRO
O15078
centrosomal protein
7.71E−03
3.24
1.43





290kDa


IGFBP3
PRO
P17936
insulin-like growth
8.01E−03
2.06
3.55





factor binding protein 3


DHRS11
PRO
A8MXC2
dehydrogenase/reductase
8.04E−03
0.67
2.07





(SDR family)





member 11


QSOX1
PRO
O00391
quiescin Q6 sulfhydryl
8.16E−03
3.04
4.86





oxidase 1


MASP1
PRO
F8W876
mannan-binding lectin
8.22E−03
5.31
8.56





serine peptidase 1





(C4/C2 activating





component of Ra-





reactive factor)


IgA_G1_S1
IgGLY
A2, G1, S1

8.36E−03
8.79
2.12


GP20HA
GLY
A2G2S[3, 6]2

8.60E−03
3.53
4.55





Analytic annotations: PRO; shot-gun proteomics, GE; gene expression, GLY; plasma glycomics, IgGLY; site-specific Ig glycosylation. Glycan annotations according to Oxford Symbol nomenclature. All N-glycans have two core GlcNAcs; F at the start of the abbreviation indicates a core a(1-6)fucose linked to inner GlcNAc; Mx, number (x) of mannose on core GlcNAcs; Ax, number of antenna (GlcNAc) on trimannosyl core; A2, biantennary with both GlcNAcs as β(1-2) linked; A3, triantennary with a GlcNAc linked β(1-2) to both mannose and a third GlcNAc linked β(1,4) to the α(1-3) linked mannose; A3′; triantennary with a GlcNAc linked β(1-2) to both mannose and the third GlcNAc linked β(1-6) mannose; B, bisecting GlcNAc linked β(1-4) to β (1-3) mannose; Gx, number (x) of β1-4 linked galasose on the antenna; Fx, number (x) of linked fucose on antenna, (4) or (3) after the F indicates that the Fuc is α(1-4) or α(1-3) linked to the GlcNAc; Sx, number (x) sialic acids linked to galactose; the number 3 or 6 in parenthesis after S indicates whether the sialic acid is in an α(2-3) or α(2-6) linkage.






OTHER EMBODIMENTS

While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features herein before set forth.


All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Claims
  • 1. A method for treating Kawasaki disease in a subject, said method comprising: (a) selecting a subject having a fever and one or more of: red eyes; a red swollen tongue;red skin on the palms of the hands and/or soles of the feet; peeling of the skin on the hands and/or feet; a rash on the main part of the body and/or in the genital area; and swollen lymph nodes;(b) obtaining a biological sample from said subject;(c) determining the level of one or more proteins from Table 1 and/or Table 2 in said biological sample;(d) comparing the level of said one or more proteins to a predetermined control value; and(e) administering a Kawasaki disease therapy to said subject if the level of said one or more proteins is indicative that said subject may benefit from a Kawasaki disease therapy;wherein an increased level of at least one protein of Table 1, as compared to a reference, and/or a decreased level of at least one protein of Table 2, as compared to a reference, is indicative that the subject may benefit from a Kawasaki disease therapy.
  • 2.-52. (canceled)
Provisional Applications (2)
Number Date Country
62067123 Oct 2014 US
61987907 May 2014 US
Continuations (1)
Number Date Country
Parent 15308114 Nov 2016 US
Child 16841575 US