Title: Biomarkers for the detection of Fronto temp oral dementia FIELD OF THE INVENTION
 The present invention relates to methods and kits for classifying an individual as being afflicted with frontotemporal dementia (FTD). The invention further relates to the discrimination between different subtypes of FTD. BACKGROUND OF THE INVENTION
 Frontotemporal dementia (FTD) is a neurodegenerative disorder that clinically presents with either behaviour and personality changes or language disturbance. It is the second most prevalent dementia of patients below age 65. The disease is often misdiagnosed in the early stage, either as a psychiatric disorder, or as a different type of dementia such as Alzheimer's disease (AD).
The underlying pathological spectrum of FTD, frontotemporal lobar degeneration (FTLD), can be divided in 2 main subtypes characterized by either tau or TDP-43 accumulations. These subtypes being referred to as FTLD-TDP-43 and FTLD-Tau. Correct diagnosis and subtyping is very relevant to determine patient management plans and to boost therapy development, especially to develop treatments targeting either Tau or TDP-43 pathological mechanisms.
Thus far, no reliable (set of) biomarker(s) with both high sensitivity and high specificity is available for FTD, let alone its pathological subtypes. The cerebrospinal fluid (CSF) biomarkers for AD, i.e. (p)Tau and amyloid beta-42, appear to be of limited value for the diagnosis of clinical FTD. While structural MM scans can be used to detect frontal lobe or temporal lobe atrophy, the early stages of FTD are normally not detectable. Therefore, novel biomarkers for diagnosis and prognosis and stratification of FTD subtypes are needed.  SUMMARY OF THE INVENTION
One aspect of the disclosure provides a method of classifying the FTD (fr onto temp oral dementia ) status of an individual,comprising determining the concentration of at least one biomarker selected from Tables 2a, 2b, 2c, 2d, or 2e from a biological sample from said individual, and determining said FTD status based on the concentration of at least one of the biomarkers selected. Preferably, at least one biomarker selected from Table 2d and at least one biomarker selected from table 2e are determined. Preferably, the individual's FTD status is classified as
a) FTD negative
b) FTD positive
c) FTD-tau positive, or
d) FTD-TDP-43 positive.
Preferably, the method comprises determining the concentration of at least one biomarker selected from table 2a, wherein the alteration in the concentration of a biomarker from table 2a as compared to a FTD negative reference value classifies the individual as FTD-tau positive. Preferably, the method comprises determining the concentration of at least one biomarker selected from tables 2b or 2c, wherein the alteration in the concentration of a biomarker from table 2b or 2c as compared to a FTD negative reference value classifies the individual as FTD positive.
Preferably, the method comprises determining the concentration of at least one biomarker selected from table 2d, preferably wherein the alteration in the
concentration of a biomarker from table 2d as compared to a FTD negative reference value classifies the individual as FTD positive. Preferably, the method further comprises determining the concentration of at least one biomarker selected from table 2e. Preferably, the method comprises determining the concentration of at least one biomarker selected from table 2e, wherein the alteration in the concentration of a biomarker from table 2e as compared to a FTD-tau positive reference value classifies the individual as FTD-TDP-43 positive.  Preferably, the sample is a bodily fluid preferably selected from blood or cerebrospinal fluid.
 Preferably, the level of said biomarkers is determined by an immunoassay.
Preferably, the level of said biomarkers is determined by mass spectrometry.
One aspect of the disclosure provides for a kit for determining the concentration of at least two biomarkers in a bodily fluid, said kit comprising two binding agents, preferably wherein the binding agents are antibodies, each binding agent directed to a different biomarker selected from Tables 2a, 2b, 2c, 2d, and 2e.
Preferably the kit comprises at least one binding agent directed to a biomarker selected from Table 2d and at least one binding agent directed to a biomarker selected from Table 2e.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 FABP4 levels in CSF of the validation cohort as measured by ELISA.
**p<0.01.
 Figure 2 Complement factor D levels in CSF of the validation cohort as measured by ELISA.
 Figure 3 YKL-40 (i.e., CHI3L1) levels in CSF of the validation cohort as measured by ELISA. Levels were significantly higher in the FTD-tau group compared to controls, TDP-43, AD, DLB and VaD. ** pO.01.
 Figure 4 MFGE-8 levels in CSF of the validation cohort as measured by ELISA. Levels were significantly decreased in FTD-TDP-43 subtype compared to control and AD. Moreover, levels were decreased in VaD patients compare to controls, AD and DBL (**p<0.01).
 Figure 5 FABP4 levels in serum of the validation cohort as measured by ELISA. Levels were lower in the FTD-TDP-43 group compared to control (p=0.05).
Figure 6 YKL-40 (i.e., CHI3L1) levels in serum of the validation cohort as measured by ELISA.
Figure 7 Decision trees for diagnosis of FTD and its subtypes.  DETAILED DESCRIPTION OF THE DISCLOSED EMBODIMENTS
The disclosure provides methods for classifying the FTD status of individual. The methods described herein can be used to determine whether an individual is afflicted with FTD, as well as to distinguish between FTD subtypes FTD-tau and FTD-TDP-43. As used herein, an individual's FTD status can be classified as
 1) FTD negative
 2) FTD positive
 3) FTD-tau positive, or
4) FTD-TDP-43 positive.
Using unbiased in-depth mass-spectrometry based proteomics, candidate biomarkers were identified that are differentially expressed in FTD compared to controls, or between FTD-TDP-43 and FTD-tau subtypes. As can be seen from Tables 2a-2e, biomarkers are identified which can diagnosis FTD generally (both forms of FTD) in individuals, while other biomarkers can distinguish between the two subtypes.
The use of the term FTD refers to a disorder caused by the degeneration of the brain's frontal lobes. Diagnostic criteria according to the Lund-Manchester criteria include insidious onset and gradual progression, early decline in social interpersonal conduct, early impairment in regulation of personal conduct, early emotional blunting, and early loss of insight. Symptoms of FTD generally appear between the ages of 45 and 65 years of age. In the methods disclosed herein, the concentration of at least one biomarker selected from tables 2a-2e is determined in a biological sample from an individual. The level of biomarker in the individual can be compared to a reference value. Preferably, the concentration of at least one biomarker selected from Tables 2a, 2b, 2c, and 2d is compared to a FTD negative reference value. As used herein, an FTD negative reference value is the concentration of a biomarker in a control (FTD negative) individual or the average concentration in a population of control individuals.
Preferably, the concentration of at least one biomarker selected from table 2e is compared to a FTD-tau positive reference value or an FTD-TDP-43 positive reference  value. As used herein, a FTD-tau positive reference value is the concentration of a biomarker in a FTD-tau positive individual or the average concentration in a population of FTD-tau positive individuals. As used herein, a FTD-TDP-43 positive reference value is the concentration of a biomarker in a FTD-TDP-43 positive individual or the average concentration in a population of FTD-TDP-43 positive individuals. A skilled person recognizes that the change of expression level which indicates a significant difference between the individual and the reference value will depend on the particular assay used. Preferably, a (positive or negative) fold change of at least 1.2 is significant.
The disclosure further provides a method for determining the concentration of at least two biomarkers selected from Tables 2a, 2b, 2c, 2d, and 2e from a biological sample from an individual, said method comprising contacting the sample with a first binding agent which recognizes a biomarker selected from Tables 2a, 2b, 2c, 2d, and 2e and a second binding agent which recognizes a different biomarker selected from Tables 2a, 2b, 2c, 2d, and 2e and determining the concentration of the at least two biomarkers from the amount of bound first and second binding agent.
As used herein, the terms individual, subject, or patient are used interchangeably and include mammals, such as primates and domesticated animals. Preferably said individual is a human. The methods disclosed herein may be used with healthy individuals, individuals suspected of suffering from FTD, or with individuals in which FTD has been diagnosed based on pathological or clinical symptoms. The disclosed methods are useful for not only for diagnosing or confirming a diagnosis of FTD but also distinguishing between pathological FTD subtypes.
In preferred embodiments, the level of biomarker is determined in vitro from a sample from an individual, preferably a biological fluid. Preferably, said sample is blood, urine, saliva, or cerebral spinal fluid (CSF), more preferably said sample is CSF.
Preferably, the methods disclosed herein determine the alteration in the
concentration of at least one biomarker from table 2a. Preferably an alteration in the concentration of at least one biomarker from table 2a as compared to a FTD negative  reference value classifies said individual as FTD-tau positive. Preferably an alteration in the concentration of at least one biomarker from table 2a as compared to an FTD- TDP43 positive reference value classifies said individual as FTD-tau positive. In preferred embodiments, the concentration of at least two biomarkers from table 2a are determined. The use of multiple biomarkers increases the confidence of classifying the FTD status. Therefore, when multiple markers are used, not all of the biomarkers need to exhibit a significant differential expression in order for a risk to be
determined. It is clear to a skilled person that the alteration in the concentration of one of the two tested biomarkers also classifies said individual as FTD-tau positive. In an exemplary embodiment, the increase in FABP4, IGFALS, NDRG4 or APOL1 as compared to FTD negative reference value indicates that the individual is FTD-tau positive, whereas a decrease in MYOC, SHBG, FCGBP indicates that the individual is FTD-tau positive. Preferably, the at least one biomarker from table 2a is selected from NDRG4 and APOLl.
Preferably, the methods disclosed herein determine the alteration in the
concentration of at least one biomarker from table 2b. Preferably an alteration in the concentration of at least one biomarker from table 2b as compared to a FTD negative reference value classifies said individual as FTD positive, more preferably as FTD-tau positive. In preferred embodiments, the concentration of at least two biomarkers from table 2b are determined, wherein an alteration in one classifies the individual. In an exemplary embodiment, the decrease in IL1RAP,0LFML1, ISLR2, CNTNAP3, TENM2, GALNS, F5, QDPR, PCMT1, CTSH, OLFML3, or NOV or an increase in MAT2A, CA1, CAT, S100A7, LCAT, FSTL5, CDH15, C ASP 14, ACAN, FAH, VCL, or LRP8 indicates that the individual is FTD positive, preferably as FTD-tau positive. Preferably, at least one biomarker from Table 2b is selected from IL1RAP, OLFML1, MAT2A, CA1, CAT, and S100A7.
Preferably, the methods disclosed herein determine the alteration in the
concentration of at least one biomarker from table 2c. Preferably an alteration in the concentration of at least one biomarker from table 2c as compared to a FTD negative reference value classifies said individual as FTD positive, more preferably as FTD- TDP-43 positive. In preferred embodiments, the concentration of at least two  biomarkers from table 2c are determined, wherein an alteration in one classifies the individual. In an exemplary embodiment, the decrease in GLA, LAMTOR2, MPZ, TMEM132B, IMPA1, DLD, GDA, or CPVL or the increase in CTSL1 or ST6GAL2 indicates that the individual is FTD positive. Preferably, the at least one biomarker from Table 2c is selected from GLA and LAMTOR2.
Preferably, the methods disclosed herein determine the alteration in the
concentration of at least one biomarker from table 2d. Preferably an alteration in the concentration of at least one biomarker from table 2d as compared to a FTD negative reference value classifies said individual as FTD positive. In preferred embodiments, the concentration of at least two biomarkers from table 2d are determined. It is clear to a skilled person that the alteration in the concentration of one of the two tested biomarkers also classifies said individual as FTD positive. In preferred embodiments, the concentration of at least two biomarkers from table 2d, preferably selected from SPTBN5, MFGE8, and CHI3L1, are determined and an alteration in the
concentration of the at least two biomarkers classifies said individual as FTD positive. A decrease in SPTBN5, RAD 23 B, AP2B1, CFD, MFGE8, or CHI3L1 indicates that the individual is FTD positive. Preferably, the biomarker from Table 2d is SPTBN5. Preferably the at least two biomarkers are SPTBN5 and MFGE8.
Preferably the at least two biomarkers are SPTBN5 and CHI3L1. Preferably the at least two biomarkers are MFGE8and CHI3L1.
Preferably, the methods disclosed herein determine the alteration in the
concentration of at least one biomarker from table 2e. Preferably an alteration in the concentration of at least one biomarker from table 2e as compared to a FTD-TDP-43 positive reference classifies said individual as FTD tau positive. Preferably an alteration in the concentration of at least one biomarker from table 2d as compared to a FTD-tau positive reference classifies said individual as FTD TDP-43 positive. In preferred embodiments, the concentration of at least two biomarkers from table 2e are determined. It is clear to a skilled person that the alteration in the concentration of one of the two tested biomarkers also classifies said individual. A decrease in KLK7, F13A1, HSPA8, FSCN1, CLIC4, or ABI3BP as compared to a FTD-TDP-43 positive reference indicates that the individual is FTD-tau positive. An increase in MOG,  HEXA, CHIDl, UBL3 as compared to a FTD-TDP-43 positive reference indicates that the individual is FTD-tau positive. Preferably, a decrease in KLK7, F13A1, HSPA8, FSCN1, CLIC4, or ABI3BP as compared to a FTD-tau positive reference indicates that the individual is FTD-TDP-43 positive. An increase in MOG, HEXA, CHIDl, UBL3 as compared to a FTD- tau positive reference indicates that the individual is FTD-TDP-43 positive. Preferably, the at least one biomarker from Table 2e is selected from KLK7, F13A1, HSPA8, and UBL3.
In preferred embodiments, the determination of biomarkers from tables 2a-2e described above are combined.
 Preferably, methods comprise 1) determining the concentration of at least one biomarker selected from table 2a as described above and 2) determining the concentration of at least one biomarker selected from table 2c as described above. Preferably, methods comprise 1) determining the concentration of at least one biomarker selected from table 2b as described above and 2) determining the concentration of at least one biomarker selected from table 2c as described above. Preferably, methods comprise 1) determining the concentration of at least one biomarker selected from table 2d as described above and 2) determining the concentration of at least one biomarker selected from table 2e or table 2a as described above.
Preferably, the methods comprise determining the concentration of SPTBN5 and KLK7, F13A1, HSPA8, or UBL3.
 Preferably, the methods comprise determining the concentration of MFGE8 and KLK7, F13A1, HSPA8, or UBL3.
 Preferably, the methods comprise determining the concentration of CHI3L1 and KLK7, F13A1, HSPA8, or UBL3.
 Preferably, the methods comprise determining the concentration of APOL1 and GLA or LAMTOR.
Preferably, the methods comprise determining the concentration of NDRG4 and GLA or LAMTOR.
 Preferably, the methods comprise determining the concentration of FABP4 and GLA or LAMTOR.  Preferably, the methods comprise determining the concentration of GLA and
IL1RAP, OLFML1, MAT2A, CA1, CAT, or S100A7.
 Preferably, the methods comprise determining the concentration of LAMTOR and IL1RAP, OLFML1, MAT2A, CA1, CAT, or S100A7.
The methods disclosed herein comprise determining the concentration of biomarkers. Any number of suitable assays known to one of skill in the art may be used to determine the level of the biomarkers disclosed herein. In preferred embodiments, a binding agent is used to bind and detect a biomarker disclosed herein. Preferably, the step of determining the concentration of a biomarker can also be expressed as determining the amount of binding of a binding agent to said biological sample from an individual, wherein said binding agent is specific for a biomarker selected from tables 2a-2e.
 Binding agents include antibodies as well as non-immunoglobulin binding agents, such as phage display-derived peptide binders, and antibody mimics, e.g., affibodies, tetranectins (CTLDs), adnectins (monobodies), anticalins, DARPins (ankyrins), avimers, iMabs, microbodies, peptide aptamers, Kunitz domains, aptamers and affilins. As used herein, the term "antibodies" include, e.g., monoclonal antibodies; polyclonal antibodies, chimeric, human, humanized antibodies; antigen-binding fragments including, but not limited to, Fab, F(ab'), F(ab')2, complementarity determining region (CDR) fragments, single-chain antibodies (scFv), bivalent single-chain antibodies, diabodies, triabodies, tetrabodies, artificial antibodies, phage display-derived antibodies, and other antigen recognizing immunoglobulin fragments. Preferably, said binding agent is an antibody.
Techniques for producing binding agents are well known in the art. For example, monoclonal antibodies can be made by the conventional method of immunization of a mammal, followed by isolation of plasma B cells producing the monoclonal antibodies of interest and fusion with a myeloma cell. Antibodies produced by other techniques such as recombinant antibodies are also encompassed in the disclosure ("Recombinant Proteins, monoclonal antibodies and therapeutic genes", 1999, eds Mountain, A., Ney,  U. and Schomburg, D., Wiley-VCH) as are polyclonal antibodies. Specific procedures for immunizing, additional immunogenic substances for boosting the immune system of the animals to be immunized and time scales for immunization are known in the art. Animals to be used for immunization include rabbits, goats, rats and chicks. The blood samples taken after sacrifice of the animals are processed according to standard procedures. Antibodies that specifically recognise the biomarkers can then be purified from the serum by known procedures, such as stepwise affinity purification. In addition, many commercially available antibodies exist, e.g., anti- FABP4 is available from, e.g., ProteinTech (15872- 1-AP) ; anti-CHI3Ll/CHI3Ll is available LifeSpan Biosciences( LS-C125352); anti-MFGE-8 is available from, e.g., Sigma (HPA002807)
Non-antibody molecules can be isolated or screened from compound libraries by conventional means. An automated system for generating and screening a compound library is described in U.S. Patents Nos. 5,901,069 and 5,463,564.
Suitable assays which utilize binding agents, generally referred to as binding or affinity assays, include, e.g., western blots, radio-immunoassay, ELISA (enzyme- linked immunosorbant assay), "sandwich" immunoassay, immunoradiometric assay, gel diffusion precipitation reaction, immunodiffusion assay, precipitation reaction, agglutination assay (e.g., gel agglutination assay, hemagglutination assay, etc.), complement fixation assay, immunofluorescence assay, protein A assay, and immunoelectrophoresis assay.
A typical ELISA assay performed in the art is described as follows. Briefly, the antigen is adsorbed to the wells of a microtiter plate. The wells are typically washed with a blocking buffer to block non-specific antibody binding and to minimize false positive results. Commonly used blocking agents are either protein solutions, such as BSA (typically used at concentrations between 1% and 5% (w/v) in PBS, pH=7.0), nonfat dry milk, or casein (the main protein component of non-fat dry milk).
After the blocking step, the wells of the microtiter plate are typically washed. The adsorbed antigen then undergoes the primary antibody incubation, after which it is typically washed again. Antibody/antigen complexes may then detected using a  secondary antibody labeled with chromogenic (e.g., horseradish peroxidase and TMB), fluorescent or chemiluminescent (e.g., alkaline phosphatase) means. The amount of color or fluorescence may be measured using a luminometer, a spectrophotometer, or other similar instruments. There are many common variations on the standard ELISA protocol, including e.g., competitive ELISA and sandwich ELISA which are all known to a skilled person.
Binding agents can also be immobilized on a solid support such as a chip or microarray. A biological sample is passed over the solid support. Bound proteins are then detected using any suitable method, such as surface plasmon resonance (SPR) (See e.g., WO 90/05305, herein incorporated by reference). High throughput protein arrays are known in the art and are also described in U.S. Publication No.
20080146459. As used herein, "solid support" means a generally or substantially planar substrate onto which an array of antigens is disposed. A solid support can be composed of any material suitable for carrying the array. Materials used to construct these solid supports need to meet several requirements, such as (1) the presence of surface groups that can be easily derivatized, (2) inertness to reagents used in the assay, (3) stability over time, and (4) compatibility with biological samples. For example, suitable materials include glass, silicon, silicon dioxide (i.e., silica), plastics, polymers, hydrophilic inorganic supports, and ceramic materials. Illustrative plastics and polymers include poly(tetrafluoroethylene), poly(vinylidenedifluoride), polystyrene, polycarbonate, polymethacrylate, and combinations thereof.
The disclosed biomarkers are polypeptide based, meaning that they are characterized by mass-to-charge ratio as determined by mass spectrometry, by the shape of their spectral peak in time-of-flight mass spectrometry and by their binding characteristics to adsorbent surfaces.. Preferably, a form of mass spectrometry is used in the disclosed methods. Examples of mass spectrometers are time-of-flight, magnetic sector, quadrupole filter, ion trap, ion cyclotron resonance, electrostatic sector analyzer and hybrids of these. In a further preferred method, the mass spectrometer is a laser desorption/ionization mass spectrometer. Mass spectrometry methods are  known to one in the art and are described in the examples herein, U.S. Patent Publication 20130122516, U.S. Pat. No. 5,719,060 (describing Surface Enhanced Laser Desorption and Ionization or SELDI) as well as Regnier et al. Clin Chem 2009 56: 165-171, which discusses approval of such diagnostics with the U.S. Food and Drug Administration.
In one embodiment, the biomarkers can be first captured on a chromatographic resin having chromatographic properties that bind the biomarkers. For example, one could capture the biomarkers on a cation exchange resin, wash the resin, elute the biomarkers and detect by MALDI (Matrix-assisted laser desorption/ionization. In another method, one could capture the biomarkers on a probe surface that comprises binding agents that bind the biomarkers, wash the surface to remove unbound material, elute the biomarkers from the surface and detect the eluted biomarkers by MALDI. For some probes, elution from the surface is not necessary and the biomarkers can be detected using mass spectrometry directly from the probe. In some embodiments, the sample is contacted with an affinity capture probe such as a ProteinChip array from Ciphergen Biosystems, Inc. The probe is washed with a buffer that will retain the biomarker while washing away unbound molecules. The biomarkers are detected by laser desorption/ionization mass spectrometry. These are just a few non-limiting examples of mass spectrometry technology known to a skilled person. Additional methods for detecting biomarkers using mass spectrometry are disclosed in U.S. Publication No. 20110129920.
The biomarkers may be detected in a gas phase ion spectrometer such as a time-of- flight mass spectrometer. The biomarkers are ionized by an ionization source such as a laser, the generated ions are collected by an ion optic assembly, and then a mass analyzer disperses and analyzes the passing ions. The detector then translates information of the detected ions into mass-to-charge ratios. Detection of a biomarker typically will involve detection of signal intensity. Thus, both the quantity and mass of the biomarker can be determined. Data generated by desorption and detection of biomarkers can be analyzed with the use of a programmable digital computer. The computer program analyzes the data to indicate the number of biomarkers detected,  and optionally the strength of the signal and the determined molecular mass for each biomarker detected.
It is clear to a skilled person that the concentration of biomarker as disclosed herein does not need to be necessarily determined in absolute amounts. The concentration of biomarker in relation to positive or negative reference values is also encompassed within the disclosure.
The disclosure also provides kits for determining the concentration of the biomarkers disclosed herein. Preferably, said kits comprise at least two, at least three, or at least four binding agents, wherein each agent binds to a different biomarker selected from tables 2a-2e. More preferably, said kits comprise at least one binding agent that binds a biomarker from table 2a and at least one binding agent that binds a biomarker from table 2c. More preferably, said kits comprise at least one binding agent that binds a biomarker from table 2d and at least one binding agent that binds a biomarker from table 2e. Preferably, the biomarkers are the preferred biomarkers described in the methods herein.
Preferably said kits comprise a solid support, such as a chip, a microtiter plate or a bead or resin comprising said binding agents. In some embodiments, the kits comprise mass spectrometry probes, such as ProteinChip™.
The kits may also provide washing solutions and/or detection reagents specific for either unbound binding agent or for said biomarkers (sandwich type assay).
A further aspect of the disclosure provides a method of treating an individual with FTD comprising a) classifying the FTD status of the individual as FTD positive, FTD- tau positive, or FTD TDP-43 positive by a method disclosed herein and b) treating said individual with a therapeutic which treats either the pathology or symptoms of FTD. Exemplary therapeutics include antidepressants for the treatment of FTD related symptoms. Being able to distinguish the underlying pathology in FTD, is crucial for targeted therapeutics. For example, an international Phase III trial is  currently being performed in FTD with LMTX, an inhibitor of tau- aggregation (TauRx Therapeutics).
Definitions
As used herein, "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition the verb "to consist" may be replaced by "to consist essentially of meaning that a compound or adjunct compound as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
The word "approximately" or "about" when used in association with a numerical value (approximately 10, about 10) preferably means that the value may be the given value of 10 more or less 1% of the value.
The term "treating" includes prophylactic and/or therapeutic treatments. The term "prophylactic or therapeutic" treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.
The invention is further explained in the following examples. These examples do not limit the scope of the invention, but merely serve to clarify the invention.  EXAMPLES
Methods:
Patient selection: Patients with established FTLD subtypes and controls were included from the biobanks of the Amsterdam Dementia Cohort and from the
Erasmus MC. FTD pathology was reviewed according to international criteria
{Cairns, 2007}. Pathological examination was performed according protocolized procedures by the Dutch brain bank, including specific immunostaining for
intranuclear / intracytoplasmatic TDP-43- pathology and tau pathology. Genetic testing was performed for mutations in the tau and progranulin genes and for the hexanucleotide repeat at C9orf72.
All subjects underwent extensive dementia screening at baseline, including physical and neurological examination, MMSE, neuropsychological investigation, EEC, MM and laboratory tests, including lumbar puncture. Dementia diagnoses were made by consensus in a multidisciplinary meeting according to standard criteria {Roman, 1993;McKeith, 2005 }. Probable AD was diagnosed according to the criteria of the National Institute of Neurological and Communicative Disorders and Stroke- Alzheimer's Disease and Related Disorders association (NINCDS-ADRDA) {McKhann, 1984}, and all patients met the core clinical NIA-AA criteria {McKhann, 2011}.
Definite FTD was diagnosed according to the criteria of Rascovsky et al Control groups of subjects with subjective memory complaints consisted of individuals who presented with cognitive complaints, but cognitive and laboratory investigations were normal and criteria for MCI, dementia or any other neurological or psychiatric disorders known to cause cognitive complaints were not met. Groups were matched for age and gender. Patient characteristics of the discovery and validation cohorts are presented in Table 1.
The study was performed according to the ethical principles of the Declaration of Helsinki and was approved by the local ethics committee. We obtained written informed consent from all subjects participating in the study.
CSF A642, total tau and p-tau were measured with commercially available ELISAs (Innotest 6-amyloid(l-42), Innotest hTAU-Ag and Innotest Phosphotau(181P);
 Innogenetics, Ghent, Belgium) on a routine basis as described before {Mulder, 2010 }.  Table 1 Characteristics of included patient populations
CSF sample preparation and gel electrophoresis
 For proteomics, CSF samples were recoded and analysed in a blinded fashion. To minimize inter-run bias, each gel contained 2 patients from each clinical group. All the samples were processed within the same batch of spin columns to avoid batch-to- batch variability. The depletion of top- 14 high abundant proteins was achieved as previously reported {Fratantoni, 2010}. Briefly, 1 mL aliquots of CSF from each patient were applied directly to the spin filters (Agilent Human 14 or Genway), following instructions from the manufacturer. Depleted CSF was further concentrated using 3kDa filters prior (Millipore) to loading the whole depleted CSF fraction on gradient gels from Invitrogen (NuPAGE 4-12% Bis-Tris gel, 1.5mmxl0 wells). The gels were then stained with Coomassie brilliant blue G-250 (Pierce, Rockford, IL).  In-Gel Digestion
 Before MS analysis, separated proteins were in-gel digested as previously described (Piersma et al., 2012). Briefly, gels were washed and dehydrated once in 50 mM ammonium bicarbonate (ABC) and twice in 50 mM ABC/50% acetonitrile (ACN). Cysteine bonds were reduced by incubation with 10 mM DTT/50 mM ABC at 56°C for 1 h and alkylated with 50 mM iodoacetamide/50 mM ABC at room temperature (RT) in the dark for 45 minutes. After washing sequentially with ABC and ABC/50% ACN, the whole gel was sliced in 5 bands for each lane. Gel parts were sliced up into approximately 1-mm cubes and collected in tubes, washed in ABC/ACN and dried in a vacuum centrifuge. Gel cubes were incubated overnight at 23°C with 6.25 ng/mL trypsin and covered with ABC to allow digestion. Peptides were extracted once in 1% formic acid and twice in 5% formic acid/50% ACN. The volume was reduced to 50 μΕ in a vacuum centrifuge prior to LC-MS analysis. NanoLC-MS/MS Analysis.
 Extracted peptides were separated on a 75 μηι x 42 cm custom packed Reprosil C18 aqua column (1.9 μηι, 120 A) in a 90 min. gradient (2-32% Acetonitrile + 0.5% Acetic acid at 300 nl/min) using a U3000 RSLC high pressure nanoLC (Dionex). Eluting peptides were measured on-line by a Q Exactive mass spectrometer (ThermoFisher Scientific) operating in data- dependent acquisition mode. Peptides were ionized using a stainless-steel emitter at a potential of +2 kV (ThermoScientific). Intact peptide ions were detected at a resolution of 35,000 (at m/z 200) and fragment ions at a resolution of 17,500 (at m/z 200); the MS mass range was 350- 1,500 Da. AGC Target settings for MS were 3E6 charges and for MS/MS 2E5 charges. Peptides were selected for Higher- energy C-trap dissociation fragmentation at an underfill ratio of 1% and a quadrupole isolation window of 1.5 Da, peptides were fragmented at a normalized collision energy of 30. Raw files from MS analysis were processed using the MaxQuant computational proteomics platform version 1.3.0.5 (Cox and Mann, 2008).
Database Searching and Statistics
MS/MS spectra were searched against the Uniprot human database (release 2013_1 January 9th, 2013 , excluding fragment, 43,688 sequences) with initial precursor and fragment mass tolerance set to 7 and 20 p. p.m., respectively. Peptides with minimum of seven amino-acid length were considered with both the peptide and protein false  discovery rate (FDR) set to 1%. Enzyme specificity was set to trypsin and up to two missed cleavage sites were allowed. Cysteine carbamidomethylation (Cys, +57.021464 Da) was searched as a fixed modification, whereas N-acetylation of proteins (N- terminal, +42.010565 Da), oxidized methionine (Met,+15.994915 Da), and serine, threonine, and tyrosine phosphorylations (Ser/Thr/Tyr, +79.966331 Da) were searched as variable modifications.
 For each identified protein, the number of spectra was exported to a spreadsheet and normalized according to previously published procedures {Pham, 2010}. The beta- binomial test was performed to identify differentially expressed proteins among the different groups {Pham, 2010}.
 Candidate biomarkers for validation were selected based on the following criteria: 1) fold change >1.2 and p-value <0.05; 2) mean spectral count of one of the clinical groups >2; 3) number of identified peptide sequences (>2?) . Next, we searched for availability of ELISA and antibodies, and selected those ELISAs with a detailed validation report (i.e. recovery, linearity results and coefficients of variation presented of individual samples) and those antibodies of which western blot reactivity against the full protein and human brain tissue was shown by the provider.
Protein expression clustering and biological functions
Functional annotation analysis has been carried out using different tools. As an initial step, all the identified proteins were searched using PANTHER classification system to retrieve a general gene ontology (GO) description of the proteomes. Then, putative biomarkers were analysed using DAVID knowledgebase {Huang, 200} to find specific enrichment of GO annotations either with respect to whole genome and the whole dataset. In particular, to reduce the redundancy of GO categories, the functional annotation clustering tool embedded in DAVID website was used. Results were reported using the most significant GO categories for each of the cluster found with the software.
 Expression data were clustered using an unsupervised approach, a K-means method implemented in-house with a MATLAB script. Protein abundances were normalized to zero, mean and unit variance per protein. The number of clusters was arbitrarily set to 16. Proteins in the expression profiles were then analysed with DAVID bioinformatics tools in order to: i) gather as much information as possible on the  biological functions of the proteins ii) see if the different expression profiles could match specific sets of biological functions, defining the possible involvement of those proteins in FTD pathological process. Assay validation
 Human Adipocyte fatty acid protein (AFABP/FABP4) ELISA kit (Cat. No
RD191036200R) obtained from BioVendor (Karasek, Czech Republic) was used to measure the concentration of fatty acid binding protein 4 (FABP4). The recombinant AFABP protein was used as standard. CHI3Llwas detected by use of the MicroVue CHI3L1 enzyme immunoassay kit (Cat. No. 8020, QUIDEL Corporation, San Diego, USA). Purified CHI3L1 from osteosarcoma MG-63 cells was used as standard.
Complement Factor D Quantikinine ELISA (Cat. No. DFD00) was from R&D Systems, Abingdon , UK.
 We tested the analytical performance of these assays for analysis of CSF. This was done with two different pools of CSF, prepared from left overs from routine diagnostics. The concentration and optimal dilution of the candidate biomarker was determined. Initial validation of linearity was performed by evaluating three serial dilutions of the CSF pools, the recovery of which had to be between 80 ad 120%, and we determined the paralellism between the standard curve and internal control samples, which also had to be between 80 and 120%. Intra-assay coefficient of variation had to be below 10%. For Western blot analysis, we analyzed whether a band was present in CSF at the expected molecular weight, if this signal was reproducible on two different days, and if the coefficient of variation between duplicate samples was acceptable.
Since the analysis of CSF from AD, LBD and VD was performed on a different day with a different batch of ELISA assays, we performed bridging by reanalysing n... samples from the validation study of FTD CSF samples.
Statistics in validation studies:
Statistics were performed in SPSS (version 20). Differences in mean values between clinical groups obtained in ELISA and Western blot experiments were analysed with with Mann-Whitney U test (2-groups), and Wilcoxon signed rank tests, applying a Bonferoni correction for multiple testing. Spearman correlation analyses were  performed to study correlations between validated biomarkers and clinical and biochemical parameters. Forward logistic regression analysis was performed, with control or FTD (both pathological subtypes) and including all validated biomarkers. A p-value smaller than 0.05 was considered significant.
Results:
 Identification of discriminatory CSF biomarkers and functions
 To facilitate in-depth coverage of the CSF proteome, CSF samples were subjected to high-abundant protein depletion followed by SDS-PAGE fractionation, in-gel tryptic digestion and nanoLC-MS/MS analysis. In total 898 proteins were identified in CSF. The beta binomial test for comparison of the protein spectral counts in the three patient groups yielded 57 differentially regulated proteins (Table 2).
FTD-tau vs both SMC and TDP-43: In total six proteins were differentially regulated between FTD-Tau patients compared to SMC and FTD-TDP-43 patients (Table 2a). The proteins MYOC, APOL1 and NDRG4 had the highest fold change (>3.5 higher spectral counts in FTD-Tau compared to both SMC and TDP-43).
FTD-tau vs SMC: In total 25 proteins were differentially regulated between FTD-Tau patients compared to SMC only (Table 2b). The proteins IL1RAP, OLFML1 (both increased), MAT2A, CA1, CAT, S100A7 and MYOC (all five decreased) had the highest fold change (<-2.0 times decreased in FTD-Tau compared to SMC and >2.5 fold increased in FTD-Tau compared to SMC).
 FTD-TDP-43 vs SMC: In total 10 CSF proteins were differentially regulated (p<0.05) between controls and FTD-TDP-43 patients (Table 2c). Of these, GLA and LAMTOR had the largest negative fold change (<-2.5 fold decrease in FTD-TDP-43 compared to SMC).
 Both FTD-Tau and FTD-TDP-43 vs SMC: In total 5 proteins were differentially regulated between controls and FTD-Tau patients, of which 16 were upregulated and 15 down regulated (Table 2d). Among these, SPTBN5 had the largest negative fold change (<-2.5 fold decrease in both FTD subtypes compared to SMC).
FTD-Tau vs FTD-TDP-43: Ten proteins were differentially regulated between FTD- TDP-43 and FTD-Tau patients. The largest negative fold change was observed for KLK7, F13A1 and HSPA8 (<-2.0 fold), and the largest positive fold change was observed for UBL3 (>2.1 fold increase in FTD-Tau compared to FTD-TDP-43).  As illustration, we defined two main scenario's or decision trees for diagnosing each of the FTD pathological subtypes, including only the candidate biomarkers with the a minimal/highest fold change of 2, indicated in bold in Table 2. Scenario 1 in Figure 7 starts with discrimination of FTD-Tau from control and TDP-43 using one of the biomarkers from Table 2a or Table 2b. Next, biomarkers from Table 2c are used to discriminate FTD-TDP-43 from controls.
 In scenario 2, FTD diagnosis is defined first, based on combinations of biomarkers from Table 2d. Next, FTD-TDP-43 and FTD-Tau are discriminated based on biomarkers from Table 2e.
Biological pathway analyses:
 To explore whether the different expression profiles were associated with specific networks and molecular and cellulite functions, the proteins in each profile were searched with the pathway tools of IPA. Distinct networks and functions were identified for each comparison. Cellular movement was the highest listed biological function for FTD-TDP-43 vs control, cellular morphology for FTD-Tau vs control, cellular death, celllar assembly and organisation and cellular function and
maintenance were shared networks. For the comparison between FTD-Tau with FTD- TDP-43, lipid metabolism was the highest list biological function, and metabolic functions were well represented.
Validation:
 Three out of six tested ELISA assays and 1 out of two Western Blot assays met our validation criteria for reliable detection in CSF. These were FABP4, CHI3L1 and complement factor D, which are all related to inflammatory function. We next continued validation of these markers in a partly independent and larger cohort (n=51) of pathologically or genetically confirmed FTD pathology patients and SMC. The results in Figure 1 show that the levels of FABP4 were increased in the group of Tau-FTD patients compared to SMC (p = 0.02) and TDP-43-FTD (p =0.04) (results corrected for age ), which extended the proteomics discovery results which showed an increase in Tau-FTD compared to controls only. The FABP4 levels were also increased 2 fold in the Tau-FTD patients compared to AD-patients (p = 0.002) and LBD-patients (p=0.001)(data corrected for age and gender). The levels of FABP4 in AD and LBD  were similar to SMC, while an increase was present in VaD compared to LBD
(p<0.01).
 The Complement factor D levels were increased in the FTD-Tau patient group compared to the two other groups, but this difference was not significant (Figure 2). The levels of the CHI3L1 were 2-fold increased in both FTD pathological subtypes compared to controls (Figure 3), which confirmed the increase in FTD-TDP-43 and FTD-Tau compared to SMC of the discovery results. Further validation in other dementia subgroups showed that the levels of this protein even higher in FTD-Tau compared to AD (P<0.002) DLB (p<0.000) and VD (p=0.006), which remained significant after correction for age and gender.
We also analysed FABP4 and CHI3L1 in paired serum of the FTD patients available in the Amsterdam cohort (n=12 FTD-TDP-43, 4 FTD-Tau, 18 SMC), but no significant differences between the groups were observed. Serum and CSF levels of FABP4 levels correlated positively (r=0.461, p=0.009), in contrast to a lack of correlation between serum and CSF for CHI3L1.
As an additional validation, the abundance of several of the markers was determined by measuring the corresponding enzyme activity. Enzyme activity was determined as follows.
Total Be a -hexosaminidase activity was measured using 3 niM 4-methylumbelliferyl- 2-acetoamido-2-deoxy-8-D-glucopyranoside (4MUGlcNAc) as substrate in 0.1 M citric acid/0.2 M disodium phosphate buffer pH 4.5. 10 μΐ of CSF were incubated with 40 μΐ of the substrate solution for 10 min at 37°C. One unit (U) of enzyme activity was defined as the amount of enzyme that hy drolyses 1 nmol of substrate/min at 37°C. Substrate specificity: 100%.
8-hexosaminidase-A was measuerd using 3 mM 4-Methylumbelliferyl 6-Sulfo-2- acetamido-2-deoxy-8-I)-glucopy.ranoside (MUGlcNA.c-6-S04) in 0.1 M citric acid/0.2 M disodium phosphate buffer pH 4.5 . 10 μΐ of CSF were incubated with 40 μΐ of the substrate solution for 60 min at 37°C. One unit (U) of enzyme activity was defined as  the amount of enzyme that hydrolyses 1 rmiol of substrate/min at 37°C. Substrate specificity: 100%,
Alpha-galactosidase A was measured using 4.5 mM 4-methylumbelliferyl- a-D- galactoside in 0.2M Na/ Acetate buffer, pH 4.5. 20 μ! of CSF were incubated with 40 μ.ί of substrate solution and 10 μΐ inhibitor alpha-galactosidase B (0.6M N-acetyl-D- galactosamine in water solution), for 120 mm at 37°C. One unit (U) of enzyme activity was defined as the amount of enzyme that hydrolyses 1 nmol of substrate/min at 37°C. Substrate specificity: 100%.
Catalase activity was analysed by a commercially available kit of Sigma, according to the manufacturer's instructions. allikrein concentrations were analysed as described by Diamandis et al, Clin Biochemistry (37:3, 2004). Specifically, CSF samples were diluted as necessary in a 60 g/1 bovine serum albumin solution and then analyzed for the following ka Slikrems, using immunofluorometric assays developed in-house: hK7, hK8 and hK10. All assays were calibrated with recombinant proteins produced in-house in yeast and
mammalian expression systems. The detection limits of these assays (in u /1) were as follows: h 7 (0.1), hK8 (0.2), and hK10 (0.05;. The within-ru and day-to-day precision of all assays was less than 10%.
Combination analysis:
We performed binary logistic regression to demonstrate the improved classification of pathological subtypes when two markers from the tables are used. Classification is expressed as sensitivity and specificity: sensitivity stands for percentage of patients that are correctly classified as patients using the test (measure for "false negatives"); specificity is the percentage of controls that are correctly classified as controls using the test (measure for "false positives").
Combination of markers from Table 2c and Table 2d:
 The use of GLA (Table 2c) as a single marker discriminated FTD positive samples (samples from patients that were clinically defined as FTD) from FTD negative controls with 80% sensitivity and 80% specificity.  In regards to discriminating between FTD subtypes, thus TDP-43-FTD from Tau- FTD, the percentages for sensitivity and specificity were as follows:
73% of clinically defined FTD-TDP-43 positive samples were correctly identified as TDP-43 positive (i.e, not Tau),and 50% of Tau positive samples were correctly identified as tau positive (i.e., not TDP-43) (Overall 67%)
For improving the ability to discriminate between FTD subtypes, thus TDP-43-FTD from Tau-FTD, addition of CHI311 (Table 2d) to GLA demonstrated added value. Including both GLA (Table 2c) and CHI3L1 (Table 2d) as biomarkers resulted in the following percentages for sensitivity and specificity:
 82% of clinically defined FTD-TDP-43 positive samples were correctly identified as TDP-43 positive and 75% of Tau positive samples were correctly identified as Tau positive. Thus, overall 80% correctly classified both FTD-TDP-43 and FTD-Tau. Thus, the combination of markers from tables 2c and 2d improved the classification between the FTD subtypes and between these subtypes and controls.
Combination of markers from Table 2d and Table 2e
 The use of MFGE8 (Table 2d) as a single marker discriminated FTD positive samples (samples from patients that were clinically defined as FTD) from FTD negative controls with 81% sensitivity and 71% specificity.
In regards to discriminating between FTD subtypes, thus TDP-43-FTD from Tau- FTD, the percentages for sensitivity and specificity were as follows:
To specifically discriminate FTD-TDP-43 from controls, use of MFGE8, 64% of clinically defined FTD-TDP-43 positive samples were correctly identified as TDP-43 positive (i.e. not control), and 77% of FTD -negative control samples were correctly identified as controls. Thus, overall 71% correctly classified both FTD-TDP-43 and controls.
For improving the ability to specifically identify TDP-43-FTD from control addition of HexA (Table 2e) to MFGE-8 demonstrated added value.  Including both HexA (Table 2e) and MFGE-8 (Table 2d) as biomarkers resulted in the following percentages for sensitivity and specificity:
 82% of clinically defined FTD-TDP-43 positive samples were correctly identified as TDP-43 positive and 73% of control samples were correctly identified as controls. Thus, overall 77% correctly classified both FTD-TDP-43 and controls.
Thus, the combination of markers from tables 2d and 2e improved the classification between the FTD subtypes and between these subtypes and controls. Combination of two markers from Table 2d
 The use of CHI3L1 (Table 2d) as a single marker discriminated FTD positive samples (samples from patients that were clinically defined as FTD) from FTD negative controls with 69% sensitivity and 65% specificity. Including both CHI3L1 (Table 2d) and MFGE-8 (Table 2d) as biomarkers resulted in the following percentages for sensitivity and specificity:
 75% of clinically defined FTD (either FTD-TDP-43 or FTD-tau) positive samples were correctly identified as FTD positive and 88% of FTD -negative control samples were correctly identified as controls. Thus, overall 82% correctly classified both FTD and controls.
Table 2a FTD-tau discriminated from control and TDP-43
 Control
 control Tau vs
 Gene Name vs. TDP- vs tau TDP-43
 43
 Fold Fold Fold
 p-value p-value p-value change change change
 MYOC -1,817 0,011 2,054 0,016
SHBG -1,420 0,026 1,527 0,008
FCGBP -1,335 0,002 1,427 0,049
IGFALS 1,267 0,005 -1,299 0,011
NDRG4 3,459 0,000 -2,729 0,000  APOL1 5,127 0,001 -2,595 0,009
FABP4* 1.710 0,047 -1,500 0,039
* validated from immunoassay; increase in FTD-tau over control and FTD-TDP-43 Table 2b FTD-tau discriminated from control.
 Control
 Gene control vs vs. TDP- Tau vs TDP- Name tau 43 43
 Fold Fold Fold
 change p-value change p-value change p-value
IL1RAP -2,533 0,034
OLFML1 -2,013 0,023
MAT2A 2,471 0,022
CA1 3,283 0,036
CAT# 4,473 0,031
S100A7 4,591 0,007
ISL 2 -1,969 0,014
CNTNAP3 -1,850 0,039
TENM2 -1,655 0,012
GALNS -1,462 0,041
F5 -1,424 0,037
Q.DPR -1,368 0,028
PCMT1 -1,307 0,049
CTSH -1,307 0,018
OLFML3 -1,277 0,027
NOV -1,266 0,011
LCAT 1,347 0,032  FSTL5 1,406 0,045
 CDH15 1,519 0,019
 CAS P 14 1,660 0,043
 ACAN 1,671 0,026
 FAH 1,731 0,018
 VCL 1,856 0,036
 L P8 1,901 0,013
# validated in enzyme assay
 Table 2c FTD-TDP-43 discriminated from control
# validated in enzyme assay  Table 2d FTD discriminated from control
 validated from immunoassay; increase in FTD-tau over control and FTD-TDP-43 Table 2e TDP-43 discriminated from Tau.
 Control
 Gene control vs Tau vs
 vs. TDP- Name tau TDP-43
 43
 Fold Fold Fold
 p-value p-value p-value change change change
 KLK7 -2,064 0,029
F13A1 -2,003 0,026
HSPA8 -1,634 0,036
UBL3 2,168 0,045
FSCN1 -1,491 0,032
CLIC4 -1,481 0,030
ABI3BP -1,268 0,044
MOG 1,295 0,042  H EXA# 1,309 0,009
CH ID1 1,553 0,035
# validated in enzyme assay
Table 3 provides the protein identities, gene and protein names and the sequences of the peptides identified in the examples. It is clear to a skilled person that determining the concentration of the biomarkers listed in the tables 2a- 2e includes determining the concentration of fragments of said biomarkers, such as described in the examples. Preferred fragments of the biomarkers are disclosed in Table 3.
Table 3
Prote Gene Protein name Identified Sequence Seq ID
D3YT ABI3B Target of Nesh-SH3 DAIWTERPFNSDSYSECK 1
 FKGPHVR 2
FVGVQLCNSLR 3
FYNIGDQR 4
GHGEDHCQFVDSFLDGR 5
IYLSDSLTGK 6
KFVGVQLCNSLR 7
NPLGEGPVSNTVAFSTESADPR 8
RPPLPPRPTHPR 9
TGQQLTSDQLPIK 10
TGQQLTSDQLPIKEGYFR 1 1
VSEPVSAGR 12
YIQKPDNSPCSITDSVK 13
E7EX ACAN AISTRYTLDFDR 14
 ARPNCGGNLLGVR 15
CYAGWLADGSLR 16
DRYEINSLVR 17
EGCYGDKDEFPGVR 18
EKEVVLLVATEGR 19
EQQSHLSSIVTPEEQEFVNNNAQDY 20
EVVLLVATEGR 21
FTFQEAANECR 22
GEWNDVPCNYHLPFTCK 23
GIVFHYR 24
GTVACGEPPVVEHAR 25
WWTQAQAH D L V I K 53
P0091 CA1 Carbonic anhydrase 1 ADGLAVIGVLMK 54
 ASPDWGYDDK 55
ASPDWGYDDKNGPEQWSK 56
EIINVGHSFHVNFEDNDNR 57
ESISVSSEQLAQFR 58
GGPFSDSYR 59
HDTSLKPISVSYNPATAK 60
LFQFHFHWGSTNEHGSEHTVDGVK 61
LQKVLDALQAIK 62
LYPIANGNNQSPVDIK 63
MASPDWGYDDK 64
SLLSNVEGDNAVPMQHNNRPTQPLK 65
TSETKHDTSLKPISVSYNPATAK 66
VGEANPKLQK 67
VLDALQAIK 68
YSAELHVAHWNSAK 69
YSSLAEAASK 70
P319 CASP Caspase-14;Caspase-14 subunit DPTAEQFQEELEK 71
 EDPVSCAFVVLMAHGR 72
EGSEEDLDALEHMFR 73
FQQAIDSR 74
KTNPEIQSTLR 75
KTNPEIQSTLRK 76
LENLFEALNNK 77
MAEAELVQEGK 78
RDPTAEQFQEELEK 79
RMAEAELVQEGK 80
TNPEIQSTLR 81
VYIIQACR 82
P040 CAT Catalase ADSRDPASDQMQHWK 83
 ADVLTTGAGNPVGDK 84
ADVLTTGAGNPVGDKLNVITVGPR 85
AFYVNVLNEEQR 86
DAQIFIQK 87
DLFNAIATGK 88
DPILFPSFIHSQK 89
FNTANDDNVTQVR 90
FSTVAGESGSADTVR 91
FSTVAGESGSADTVRDPR 92
GAGAFGYFEVTHDITK 93
GPLLVQDVVFTDEMAHFDR 94
HMNGYGSHTFK 95
LCENIAGHLK 96
LCENIAGHLKDAQIFIQK 97
LFAYPDTHR 98
LGPNYLHIPVNCPYR 99
LSQEDPDYGIR 100
LVNANGEAVYCK 101
NAIHTFVQSGSHLAAR 102
NFTEVHPDYGSHIQALLDK 103
NFTEVHPDYGSHIQALLDKYNAEKPK 104
NLSVEDAAR 105
RFNTANDDNVTQVR 106
VWPHKDYPLIPVGK 107
P552 CDH1 Cadherin-15 AEATDADDPETDNAALR 108
 AIVLAQDDASQPR 109
ALDYESCEHYELK 1 10
AWVIPPISVSENHK 1 1 1
AWVIPPISVSENHKR 1 12
 DLPGSPNWVAR 1 13
DPDTEQLQR 1 14
DYDPEDWLQVDAATGR 1 15
FTILEGDPDGQFTIR 1 16
GVFSIDK 1 17
GVFSIDKFTGK 1 18
HQVPEGLHR 1 19
IQTQHVLSPASPFLK 120
LEVEDRDLPGSPNWVAR 121
LPYPLVQIK 122
SDKQQLGSVIYSIQGPGVDEEPR 123
TNEGVLSIVK 124
TSLAEGAPPGTLVATFSAR 125
VFLNAMLDR 126
VHVQDTNEPPVFQENPLR 127
VLEGAVPGTYVTR 128
VSVQNEAPLQAAALR 129
K7ER CFD Complement factor D ATLGPAVR 130
 ATLGPAVRPLPWQR 131
AVPHPDSQPDTIDHDLLLLQLSEK 132
DSCKGDSGGPLVCGGVLEGVVTSGS 133
DVAPGTLCDVAGWGIVNHAGR 134
GDSGGPLVCGGVLEGVVTSGSR 135
KKPGIYTR 136
LMCAESNRR 137
RDSCKGDSGGPLVCGGVLEGVVTS 138
RLYDVLR 139
RPDSLQHVLLPVLDR 140
RTHHDGAITER 141
VASYAAW I DSVLA 142
 VDRDVAPGTLCDVAGWGIVNHAGR 143
VQVLLGAHSLSQPEPSK 144
VQVLLGAHSLSQPEPSKR 145
P362 CHI3L C itinase-3-like protein 1 AEFIKEAQPGK 146
 AEFIKEAQPGKK 147
DKQHFTTLIK 148
DKQHFTTLIKEMK 149
EAGTLAYYEICDFLR 150
EGDGSCFPDALDR 151
EMKAEFIK 152
FPLTNAIK 153
FPLTNAIKDALAAT 154
FSKIASNTQSR 155
FSNTDYAVGYMLR 156
FTKEAGTLAYYEICDFLR 157
GNQWVGYDDQESVK 158
GNQWVGYDDQESVKSK 159
GQEDASPDRFSNTDYAVGYMLR 160
GQEDASPDRFSNTDYAVGYMLRLGA 161
GTTGHHSPLFR 162
ILGQQVPYATK 163
ILGQQVPYATKGNQWVGYDDQESVK 164
ISQHLDFISIMTYDFHGAWR 165
KQLLLSAALSAGK 166
LGAPASKLVMG I PTFG R 167
LVCYYTSWSQYR 168
LVMGIPTFGR 169
LVMGIPTFGRSFTLASSETGVGAPIS 170
NRNPNLKTLLSVGGWNFGSQR 171
 QHFTTLIK 172
QLAGAMVWALDLDDFQGSFCGQDL 173
QLLLSAALSAGK 174
RTFIKSVPPFLR 175
SFTLASSETGVGAPISGPGIPGR 176
SFTLASSETGVGAPISGPGIPGRFTK 177
SKVQYLK 178
SKVQYLKDR 179
SVPPFLR 180
TFIKSVPPFLR 181
THGFDGLDLAWLYPGR 182
THGFDGLDLAWLYPGRR 183
TLLSVGGWNFGSQR 184
TLLSVGGWNFGSQRFSKIASNTQSR 185
VQYLKDR 186
VTIDSSYDIAK 187
J3KN CHID Chitinase domain-containing protein 1 DAREPVVGAR 188
 DRHFAGDVLGYVTPWNSHGYDVTK 189
EMFEVTGLHDVDQGWMR 190
FTQISPVWLQLK 191
GLVVTDLK 192
GLVVTDLKAESVVLEHR 193
HVVFYPTLK 194
LLFEDWTYDDFR 195
MVWDSQASEHFFEYK 196
NVLDSEDEIEELSK 197
SQFSDKPVQDR 198
Q9Y6 CLIC4 Chloride intracellular channel protein 4 AGSDGESIGNCPFSQR 199
 DEFTNTCPSDKEVEIAYSDVAK 200
EMTGIWR 201
 GVVFSVTTVDLK 202
GVVFSVTTVDLKR 203
HPESNTAGMDIFAK 204
KFLDGNEMTLADCNLLPK 205
KPADLQNLAPGTHPPFITFNSEVK 206
NSRPEANEALER 207
YLTNAYSR 208
Q9BZ CNTN Contactin-associated protein-like 3 LSLFQPGQSPR 209
 SAVNLAER 210
Q9H3 CPVL Probable serine carboxypeptidase CPVL AFDMINR 21 1
 CTEPEDQLYYVK 212
EDTVQSVKPWLTEIMNNYK 213
ELSLVGPFPGLNMK 214
FLSLPEVR 215
GDSGQPLFLTPYIEAGK 216
GGGHILPYDQPLR 217
GRELSLVGPFPGLNMK 218
GWDPYVG 219
IFKSDSEVAGYIR 220
KQNWFEAFEILDK 221
NNDFYVTGESYAGK 222
QAGDFHQVIIR 223
QCHECIEHIR 224
QCHECIEHIRK 225
QNWFEAFEILDK 226
SDSEVAGYIR 227
SLMGMDWK 228
SVSMPPKGDSGQPLFLTPYIEAGK 229
YLREDTVQSVKPWLTEIMNNYK 230
 YVPAIAHLIHSLNPVR 231
P096 CTSH Pro-cathepsin H;Cathepsin H mini G I MG E DTYPYQG K 232
 G I MG E DTYPYQG KDGYCK 233
GKNMCGLAACASYPIPLV 234
GNFVSPVK 235
GTGPYPPSVDWR 236
KGNFVSPVK 237
KKGNFVSPVK 238
KTYSTEEYHHR 239
LQTFASNWR 240
MALNQFSDMSFAEIK 241
MLSLAEQQLVDCAQDFNNHGCQGG 242
NGIPYWIVK 243
NMCGLAACASYPIPLV 244
NSWGPQWGMNGYFLIER 245
TGIYSSTSCHK 246
TPDKVNHAVLAVGYGEK 247
TYSTEEYHHR 248
VNHAVLAVGYGEK 249
P077 CTSL Cathepsin L1 ;Cathepsin L1 heavy AVATVGPISVAIDAGHESFLFYK 250
 GKVFQEPLFYEAPR 251
HSFTMAMNAFGDMTSEEFR 252
LYGMNEEGWR 253
LYGMNEEGWRR 254
MIELHNQEYR 255
NHCGIASAASYPTV 256
NQGQCGSCWAFSATGALEGQMFR 257
NSWGEEWGMGGYVK 258
QVMNGFQNR 259
RNHCGIASAASYPTV 260
 VFQEPLFYEAPR 261
P096 DLD Dihydrolipoyl dehydrogenase, ADGGTQVIDTK 262
 ALLNNSHYYHMAHGK 263
ALTGGIAHLFK 264
EANLAASFGK 265
GIEMSEVR 266
GRIPVNTR 267
IPNIYAIGDVVAGPMLAHK 268
NETLGGTCLNVGCIPSK 269
NLGLEELGIELDPR 270
SEEQLKEEGIEYK 271
VCHAHPTLSEAFR 272
VGKFPFAANSR 273
P004 F13A Coagulation factor XIII A chain EIRPNSTVQWEEVCRPWVSGHR 274
 ETFDVTLEPLSFK 275
GVNLQEFLNVTSVHLFK 276
KDGTHVVENVDATHIGK 277
KPLNTEGVMK 278
LALETALMYGAK 279
MYVAVWTPYGVLR 280
STVLTIPEIIIK 281
VEYVIGR 282
WDTNKVDHHTDKYENNK 283
P122 F5 Coagulation factor V;Coagulation factor V ADKPLSIHPQGIR 284
 AGMQTPFLIMDR 285
AGMQTPFLIMDRDCR 286
APSHQQATTAGSPLR 287
ASEFLGYWEPR 288
ASKPGWWLLNTEVGENQR 289
AVQPGETYTYK 290
AWAYYSAVNPEKDIHSGLIGPLLICQK 291
AWGESTPLANKPGK 292
DGTDYIEIIPK 293
DIHSGLIGPLLICQK 294
DPDNIAAWYLR 295
DPPSDLLLLK 296
DSNMPMDMR 297
EDGILGPIIR 298
EEVQSSEDDYAEIDYVPYDDPYK 299
EFNPLVIVGLSK 300
EFVLLFMTFDEK 301
EFVLLFMTFDEKK 302
EKHTHHAPLSPR 303
EKPQSTISGLLGPTLYAEVGDIIK 304
ENQFDPPIVAR 305
ETDIEDSDDIPEDTTYK 306
ETDIEDSDDIPEDTTYKK 307
EYEPYFK 308
EYEPYFKK 309
FCENPDEVK 310
FCENPDEVKR 31 1
FCENPDEVKRDDPK 312
FTVNNLAEPQK 313
GEYEEHLGILGPIIR 314
HEDTLTLFPMR 315
HLQAGMQAYIDIK 316
HLSQDTGSPSGMRPWEDLPSQDTG 317
HSLVLHK 318
HTHHAPLSPR 319
HTVNPNMKEDGILGPIIR 320
IFEGNTNTK 321
ITAIITQGCK 322
IVYREYEPYFKK 323
KITAIITQGCK 324
KMHDRLEPEDEESDADYDYQNR 325
KSHEFHAINGMIYSLPGLK 326
KSWWGDYWEPFR 327
KSWYYEK 328
KVMYTQYEDESFTK 329
LAAALGIR 330
LAAEFASK 331
LAAEFASKPWIQVDMQK 332
LELFGCDIY 333
LELQGCEVNGCSTPLGMENGK 334
LELQGCEVNGCSTPLGMENGKIENK 335
LHLLNIGGSQDIHVVHFHGQTLLENG 336
LKHSLVLHK 337
LLSLGAGEFK 338
LNNGGSYNAWSVEK 339
LSEGASYLDHTFPAEK 340
LSEGASYLDHTFPAEKMDDAVAPGR 341
MDDAVAPGR 342
MPMGLSTGIISDSQIK 343
MRPWKDPPSDLLLLK 344
NKADKPLSIHPQGIR 345
NVMYFNGNSDASTIK 346
NVMYFNGNSDASTIKENQFDPPIVAR 347
NYYIAAEEISWDYSEFVQFt 348
 PGWWLLNTEVGENQR 349
PWIQVDMQK 350
PYYSDVDIMR 351
QHQLGVWPLLPGSFK 352
QWLEIDLLK 353
RHEDTLTLFPMR 354
SEAYNTFSER 355
SHEFHAINGMIYSLPGLK 356
SQHLDNFSNQIGK 357
SSMVDKIFEGNTNTK 358
SSSPELSEMLEYDR 359
SWWGDYWEPFR 360
SWYLEDNINK 361
SWYLEDNINKFCENPDEVKR 362
SYTIHYSEQGVEWKPYR 363
VMYTQYEDESFTK 364
WHLASEK 365
WIISSLTPK 366
WNILEFDEPTENDAQCLTR 367
WNILEFDEPTENDAQCLTRPYYSDVD 368
YLDSTFTKR 369
P150 FABP Fatty acid-binding protein, adipocyte CDAFVGTWK 370
 EDDKLVVECVMK 371
EVGVGFATR 372
LVSSENFDDYIVIKEVGVGFATR 373
NTEISFILGQEFDEVTADDRK 374
P169 FAH Fumarylacetoacetase HLFTGPVLSK 375
Q9Y6 FCGB IgGFc-binding protein AGCVAESTAVCR 376
 AGVQVWLGANGK 377
AIGYATAADCGR 378
AISGLTIDGHAVGAK 379
ALAS YVAACQAAG V V 1 E DW R 380
APGWDPLCWDECR 381
AQDFSPCYG 382
ASQHGSDVVIETDFGLR 383
AVGGKPAGWQVGGAQGCGECVSK 384
AYSHSVSLTR 385
CECGPGGHVTCQEGAACGPHEECR 386
CECGPGGHVTCQEGAACGPHEECR 387
CFEGCECDDR 388
CFEGCECDDRFLLSQGVCIPVQDCG 389
CLANGGIHYITLDGR 390
CLLPGQSGPLCDALATYAAACQAAG 391
CPGLQNTIPWYR 392
CREGGEVSCEPSSCGPHETCR 393
CSCSSSSGLTCQAAGCPPGR 394
CSVQNGLLGCYPDR 395
CTCNGATHQVTCR 396
DPCHGVTCRPQETCK 397
EEFCGLLSSPTGPLSSCHK 398
EGCVCDAGFVLSGDTCVPVGQCGCL 399
EGGEVSCEPSSCGPHETCR 400
FAVLQENVAWGNGR 401
FDFMGTCTYLLVGSCGQNAALPAFR 402
FDFMGTCVYVLAQTCGTRPGLHR 403
FDFQGTCNYVLATTGCPGVSTQGLT 404
FLLSQGVCIPVQDCGCTHNGR 405
FYPAGDVLR 406
GATTSPGVYELSSR 407
GCGEGCGPQGCPVCLAEETAPYES 408
GCVLDVCMGGGDR 409
GCVLDVCMGGGDRDILCK 410
GDKAFLCR 41 1
GEVGFVLVDNQR 412
GGGQAANALAFGNSWQEETRPGCG 413
GLQAGDVVEFEVR 414
GMVCQEHSCKPGQVCQPSGGILSCV 415
GNPAVSYVR 416
GSCPTCPEDRLEQYEGPGFCGPLAP 417
GVWVNGLR 418
GVWVNGLRVDLPAEK 419
HTTCNHVVEQLLPTSAWGTHYVVPT 420
KFDFQGTCNYVLATTGCPGVSTQGL 421
KYQKEEFCGLLSSPTGPLSSCHK 422
LASVSVSR 423
LCGACGNFDGDQTNDWHDSQEKPA 424
LCGMLTK 425
LDDGDYLCEDGCQNNCPACTPGQA 426
LDDGDYLCEDGCQNNCPACTPGQA 427
LDGPFAVCHDTLDPR 428
LDGPFAVCHDTLDPRPFLEQCVYDL 429
LDPQGAVR 430
LDPQGAVRDCVYDR 431
LDSLVAQQLQSK 432
LEDGVQACHATGCGR 433
LEQYEGPGFCGPLAPGTGGPFTTCH 434
LLFDGDAHLLMSIPSPFR 435
LLISSLSESPASVSILSQADNTSK 436
LLISSLSESPASVSILSQADNTSKK 437
LPVSLSEGR 438
LPVVLANGQIR 439
LRVPAAYAASLCGLCGNYNQDPADD 440
LRVPAAYAGSLCGLCGNYNQDPADD 441
LTYNHGGITGSR 442
LVDPQGPLK 443
NECGILADPK 444
NMVLQTTK 445
NPQGPFATCQAVLSPSEYFR 446
NTGREEFLTAFLQNYQLAYSK 447
PAGWQVGGAQGCGECVSK 448
PFLEQCVYDLCVVGGER 449
PGDEDFSIVLEK 450
QCVYDLCAQK 451
QCVYDLCAQKGDK 452
RCECGPGGHVTCQEGAACGPHEEC 453
RCECGPGGHVTCQEGAACGPHEEC 454
RCTCNGATHQVTCR 455
RFDFMGTCTYLLVGSCGQNAALPAF 456
RFDFMGTCVYVLAQTCGTRPGLHR 457
RPDFCPFQCPAHSHYELCGDSCPGS 458
RPDFCPLQCPAHSHYELCGDSCPVS 459
RVSYVGLVTVR 460
SLAAYTAACQAAGVAVKPWR 461
SPANCPLSCPANSR 462
SRLPVSLSEGR 463
SVPGCEGVALVVAQTK 464
SVPGCEGVALVVAQTKAISGLTIDGH 465
TCQGSCAALSGLTGCTTR 466
TDSFCPLHCPAHSHYSICTR 467
TEAVGQVHIFFQDGMVTLTPNK 468
TPDGSLLVR 469
TVLSPVEPSCEGMQCAAGQR 470
VAVIVSNDHAGK 471
VAYDLVYYVR 472
VDLPAEK 473
VDVTLPSSYHGAVCGLCGNMDR 474
VLVENEHR 475
VNGVLTALPVSVADGR 476
VPAAYAASLCGLCGNYNQDPADDLK 477
VPAAYAGSLCGLCGNYNQDPADDLK 478
VPSSYAEALCGLCGNFNGDPADDLA 479
VSYVGLVTVR 480
VTASSPVAVLSGHSCAQK 481
VTLQPYNVAQLQSSVDLSGSK 482
VTVNGVDMK 483
VTVNGVDMKLPVVLANGQIR 484
VTVPGNYYQLMCGLCGNYNGDPK 485
VTVPGNYYQQMCGLCGNYNGDPK 486
VVAEVQICHGK 487
VVTVAALGTNISIHK 489
VVTVAALGTNISIHKDEIGK 490
VVVCQEHSCKPGQVCQPSGGILSCV 491
VVVCQEHSCKPGQVCQPSGGILSCV 492
VYDLHGSCSYVLAQVCHPKPGDEDF 493
VYQSGPR 494
YDLAFVVASQATK 495
YQKEEFCGLLSSPTGPLSSCHK 496
YYPLGEVFYPGPECER 497
 YYPLGEVFYPGPECERR 498
YYPLGQTFYPGPGCDSLCR 499
Q166 FSCN Fascin KVTGTLDANR 500
 LINRPIIVFR 501
LSCFAQTVSPAEK 502
LVARPEPATGYTLEFR 503
PADEIAVDR 504
SSYDVFQLEFNDGAYNIK 505
VGKDELFALEQSCAQVVLQAANER 506
VTGTLDANR 507
WSLQSEAHR 508
YLAADKDGNVTCER 509
YLAPSGPSGTLK 510
YLKGDHAGVLK 51 1
YLTAEAFGFK 512
YSVQTADHR 513
Q8N4 FSTL Follistatin-related protein 5 AFQVIQLSLPEDQK 514
 CVWASAVNVK 515
DKFIYVAQPTLDR 516
DSLFILDGR 517
EPGVTASLR 518
ETGQAECACMDLCK 519
FGFILHKDEAALQK 520
GNTVIWVGDA 521
HLALEEFYR 522
HYKPVCGSDGEFYENHCEVHR 523
IDLETMSYIK 524
ITIVHNEDCFFKGDK 525
LLGFQDEVCPK 526
 LLVDQMFK 527
LNCEITEVEK 528
NEAGVDEDISSLFVEDSAR 529
NGIDITPK 530
NMLLDLQNQK 531
NNIILNNLDLEDINDFGDDGSLYITK 532
QIQDSGLFGQYLMTPSK 533
SHDQVWVLSWGTLEK 534
SYQPLMR 535
TSPTLQVITLASGNVPHHTIHTQPVGK 536
VDDFFIPTTTLIITHMR 537
VIQPIECEFQR 538
VLIVDVQSQK 539
VQYITIR 540
VVQAVSTDPVPVK 541
YFDADSNGLVDINELTQVIK 542
P340 GALN N-acetylgalactosamine-6-sulfatase AHFWTWTNSWENFR 543
 AIDGLNLLPTLLQGR 544
ARPNIPVYR 545
CLTPPESIPK 546
DWEMVGR 547
FPLSFASAEYQEALSR 548
GDTLMAATLGQHK 549
LMDRPIFYYR 550
LPLIFHLGR 551
NGFYTTNAHAR 552
PFLGTSQR 553
YYEEFPINLK 554
Q9Y2 GDA Guanine deaminase AVMVSNILLINK 555
AVMVSNILLINKVNEK 556
 DHLLGVSDSGK 557
DLHIQSHISENR 558
DLHIQSHISENRDEVEAVK 559
EFDAILINPK 560
ETTEESIKETER 561
EWCFKPCEIR 562
FLYLGDDR 563
FLYLGDDRNIEEVYVGGK 564
FQNIDFAEEVYTR 565
FSLSCSETLMGELGNIAK 566
FVSEMLQK 567
GASIAHCPNSNLSLSSGFLNVLEVLK 568
GTFVHSTWTCPMEVLR 569
IGLGTDVAGGYSYSMLDAIR 570
IVFLEEASQQEK 571
LATLGGSQALGLDGEIGNFEVGK 572
LATLGGSQALGLDGEIGNFEVGKEFD 573
NIEEVYVGGK 574
NLYPSYK 575
NYTSVYDK 576
RAVMVSNILLINK 577
TVMAHGCYLSAEELNVFHER 578
VCMDLNDTFPEYK 579
VCMDLNDTFPEYKETTEESIKETER 580
VKPIVTPR 581
YTFPAEHR 582
P062 GLA Alpha-galactosidase A ALLQDKDVIAINQDPLGK 583
 KLGFYEWTSR 584
NFADIDDSWK 585
 QLANYVHSK 586
QYCNHWR 587
SILDWTSFNQER 588
SYTIAVASLGK 589
TPTMGWLHWER 590
P068 HEXA Beta-hexosaminidase subunit alpha ALLSAPWYLNR 591
 ALVIGGEACMWGEYVDNTNLVPR 592
DLLFGSGSWPR 593
DLLFGSGSWPRPYLTGK 594
EDIPVNYMK 595
ELELVTK 596
EVIEYAR 597
GFGEDFK 598
GLETFSQLVWK 599
GLLLDTSR 600
GSYNPVTHIYTAQDVK 601
GVQAQPLNVGFCEQEFEQT 602
GYVVWQEVFDNK 603
GYVVWQEVFDNKVK 604
HYLPLSSILDTLDVMAYNK 605
IQPDTIIQVWR 606
IQPDTIIQVWREDIPVNYMK 607
ISYGPDWK 608
KGSYNPVTHIYTAQDVK 609
LNVFHWHLVDDPSFPYESFTFPELM 610
LTSDLTFAYER 61 1
LWFSLLLAAAFAGR 612
SNPEIQDFMR 613
TEIEDFPR 614
LEYLLLSR 644
 LFQGLGK 645
LFQGLGKLEYLLLSR 646
LHSLHLEGSCLGR 647
LSHNAIASLRPR 648
LWLEGNPWDCGCPLK 649
NLIAAVAPGAFLGLK 650
NLPEQVFR 651
SFEGLGQLEVLTLDHNQLQEVK 652
SLALGTFAHTPALASLGLSNNR 653
TFKDLHFLEELQLGHNR 654
TFTPQPPGLER 656
VAGLLEDTFPGLLGLR 657
WLDLSHNR 658
Q9N IL1 RA lnterleukin-1 receptor accessory protein CDDWGLDTMR 659
 DLEEPINFR 660
DSCFNSPMK 661
IKCPLFEHFLK 662
NAVPPVIHSPNDHVVYEK 663
NEVWWTIDGK 664
QIQVFEDEPAR 665
TQILSIK 666
P292 IMPA Inositol monophosphatase 1 EIQVIPLQR 667
 EIQVIPLQRDDED 668
EKYPSHSFIGEESVAAGEK 669
LFCIPVHGIR 670
LQVSQQEDITK 671
MLISSIK 672
MVLSNMEK 673
SILTDNPTWIIDPIDGTTNFVHR 674
SLLVTELGSSR 675
 SSPVDLVTATDQK 676
Q6U ISLR2 Immunoglobulin superfamily containing AGLAFVLHCIADGHPTPR 677
 DALGALPDLR 678
GAFADVTQVTSLWLAHNEVR 679
HAPGAGGEPDGQAPTSER 680
LPALPCAPPSVHLSAEPPLEAPGTPL 681
NLDLSHNFISSFPWSDLR 682
TVEPGALAVLSQLK 683
VAVAATGPPK 684
VSLPEPDSIACASPPALQGVPVYR 685
YAHQFADCAYK 686
P498 KLK7 Kallikrein-7 HPGYSTQTHVNDLMLVK 687
 LISPQDCTK 688
MNEYTVHLGSDTLGDRR 689
NACNGDSGGPLVCR 690
WINDTMK 691
WVLTAAHCK 692
Q9Y2 LAMT Ragulator complex protein LAMTOR2 ETVGFGMLK 693
 FILMDCMEGR 694
NGNQAFNEDNLK 695
VANLLLCMYAK 696
P041 LCAT Phosphatidylcholine-sterol acyltransferase FIDGFISLGAPWGGSIK 697
 ITTTSPWMFPSR 698
LAGYLHTLVQNLVNNGYVR 699
LAGYLHTLVQNLVNNGYVRDETVR 700
LDKPDVVNWMCYR 701
LEPGQQEEYYR 702
LEPGQQEEYYRK 703
PMLVLASGDNQGIPIMSSIK 704
 PVILVPGCLGNQLEAK 705
SSGLVSNAPGVQIR 706
STELCGLWQGR 707
TYIYDHGFPYTDPVGVLYEDGDDTVA 708
TYSVEYLDSSK 709
Q141 LRP8 Low-density lipoprotein receptor-related AIAVDPLR 710
 CGGDGGGACIPER 71 1
DCEKDQFQCR 712
GDEFQCGDGTCVLAIK 713
IGFECTCPAGFQLLDQK 714
IYWCDLSYR 715
LHQLSSIDFSGGNRK 716
SGECVHLGWR 717
SPSLIFTNR 718
TISVATVDGGR 719
TLISSTDFLSHPFGIAVFEDK 720
VFWTDLENEAIFSANR 721
WKCDGEEECPDGSDESEATCTK 722
P31 1 MAT2 S-adenosylmethionine synthase isoform FVIGGPQGDAGLTGR 723
 GAVLPIR 724
NFDLRPGVIVR 725
YLDEDTIYHLQPSGR 726
Q084 MFGE Lactadherin;Lactadherin short form;Medin EFVGNWNK 727
 EVTGIITQGAR 728
HKEFVGNWNK 729
ILPVAWHNR 730
LYPTSCHTACTLR 731
MWVTGVVTQGASR 732
NAVHVNLFETPVEAQYVR 733
NFGSVQFVASYK 734
 NLFETPILAR 735
TWGLHLFSWNPSYAR 736
VAYSLNGHEFDFIHDVNK 737
VAYSLNGHEFDFIHDVNKK 738
VTFLGLQHWVPELAR 739
BOUZ MOG Myelin-oligodendrocyte glycoprotein ALVGDEVELPCR 740
 DHSYQEEAAMELK 741
DQDGDQAPEYR 742
FSDEGGFTCFFR 743
GRTELLK 744
GRTELLKDAIGEGK 745
NGKDQDGDQAPEYR 746
TELLKDAIGEGK 747
P251 MPZ Myelin protein PO DAISIFHYAK 748
 GQPYIDEVGTFK 749
NPPDIVGK 750
TSQVTLYVFEK 751
YQPEGGRDAISIFHYAK 752
Q999 MYO Myocilin DPKPTYPYTQETTWR 753
 ELETAYSNLLR 754
ESPSGYLR 755
LRQENENLAR 756
LSSLESLLHQLTLDQAARPQETQEGL 757
RLESSSQEVAR 758
TLTIPFK 759
YELNTETVK 760
YSSMIDYNPLEK 761
Q9UL NDR Protein NDRG4 DLDINRPGTVPNAK 762
FPEEKPLLR 763
 GNRPAILTYHDVGLNHK 764
GWIDWAATK 765
LDPTTTTFLK 766
MADSGGLPQVTQPGK 767
MAGLQELR 768
YFLQGMGYMPSASMTRLAR 769
P487 NOV Protein NOV homolog AVLDGCSCCLVCAR 770
 CPATPPTCAPGVR 771
CQLDVLLPEPNCPAPR 772
DGQIGCVPR 773
FCGVCSDGR 774
FQCTCRDGQIGCVPR 775
GESCSDLEPCDESSGLYCDR 776
KPVMVIGTCTCHTNCPK 777
KVEVPGECCEK 778
LCMVRPCEQEPEQPTDK 779
LCMVRPCEQEPEQPTDKK 780
NNEAFLQELELK 781
QRGESCSDLEPCDESSGLYCDR 782
SADPSNQTGICTAVEGDNCVFDGVIY 783
TIQAEFQCSPGQIVK 784
VEVPGECCEK 785
Q6U OLFM Olfactomedin-like protein 1 AFMEDNTKPAPR 786
 EIDYIQYLR 787
GFLFFHNQATSNEIIK 788
IEPGTLGVEHSWDTPCR 789
SAVGNLALR 790
SHSMIHYNPR 791
VLEQGLEK 792
VYLLIGSR 793
Q9N OLFM Olfactomedin-like protein 3 AALPYFPR 794
 DFTLAMAAR 795
EALRTEADTISGR 796
EDDRHLCLAK 797
EVDYLETQNPALPCVEFDEK 798
FGGPAGLWTK 799
HAAELRDFK 800
IQCSFDASGTLTPER 801
LAALEER 802
LDPQTLDTEQQWDTPCPR 803
LRDFTLAMAAR 804
MGPSTPLLILFLLSWSGPLQGQQHHL 805
MGPSTPLLILFLLSWSGPLQGQQHHL 806
MLPLLEVAEK 807
MLPLLEVAEKER 808
NEKYDMVTDCGYTISQVR 809
QLYAWDDGYQIVYK 810
R FGGPAGLWTK 81 1
RLAALEER 812
RPPGRPGGGGEMENTLQLIK 813
YDMVTDCGYTISQVR 814
YGAHASLR 815
H7BY PCMT Protein-L-isoaspartate 0- ALDVGSGSGILTACFAR 816
 DDPTLLSSGR 817
ELVDDSVNNVR 818
KDDPTLLSSGR 819
LILPVGPAGGNQMLEQYDK 820
LILPVGPAGGNQMLEQYDKLQDGSIK 821
IVIGYAEEAPYDAIHVGAAAPVVPQALI 822
 MKPLMGVIYVPLTDK 823
MKPLMGVIYVPLTDKEK 824
SGGASHSELIHNLR 825
SGGASHSELIHNLRK 826
TDKVFEVMLATDR 827
VFEVMLATDR 828
VIGIDHIK 829
VIGIDHIKELVDDSVNNVR 830
VQLVVGDGR 831
P094 QDPR Dihydropteridine reductase AAAAAAGEAR 832
 AALDGTPGMIGYGMAK 833
EGGLLTLAGAK 834
GAVHQLCQSLAGK 835
HLKEGGLLTLAGAK 836
MTDSFTEQADQVTAEVGK 837
NCDLMWK 838
NRPSSGSLIQVVTTEGR 839
NSGMPPGAAAIAVLPVTLDTPMNR 840
NSGMPPGAAAIAVLPVTLDTPMNRK 841
QSIWTSTISSHLATK 842
RVLVYGGR 843
TELTPAYF 844
VDAILCVAGGWAGGNAK 845
VLVYGGR 846
P547 RAD2 UV excision repair protein RAD23 homolog GKDAFPVAGQK 847
 IDIDPEETVK 848
ILNDDTALK 849
ILNDDTALKEYK 850
NFVVVMVTKPK 851
P31 1 S100 Protein S100-A7 GTNYLADVFEK 852
 GTNYLADVFEKK 853
KGTNYLADVFEK 854
P070 SERP Glia-derived nexin AKIEVSEDGTK 855
 ASAATTAILIAR 856
FTAVAQTDLK 857
FTAVAQTDLKEPLK 858
HNPTGAVLFMGQINKP 859
LVLVNAVYFK 860
NKDIVTVANAVFVK 861
NKDVFQCEVR 862
SENLHVSHILQK 863
SYQVPMLAQLSVFR 864
TIDSWMSIMVPK 865
VLGITDMFDSSK 866
VQVILPK 867
P042 SHBG Sex hormone-binding globulin DDWFMLGLR 868
 DGRPEIQLHNHWAQLTVGAGPR 869
DIPQPHAEPWAFSLDLGLK 870
DS W L D KQ A E I S AS A PTS L R 871
IALGGLLFPASNLR 872
LDVDQALNR 873
LPLVPALDGCLR 874
QAAGSGHLLALGTPENPSWLSLHLQ 875
QAEISASAPTSLR 876
SCDVESNPGIFLPPGTQAEFNLR 877
TSSSFEVR 878
TWDPEGVIFYGDTNPK 879
WHQVEVK 880
Q9N SPTB Spectrin beta chain, brain 4 AALRESFLK 881
 EPVPPGDLR 882
KAALRESFLK 883
LLSTLGPR 884
SIMTYVSLYYHYCSR 885
Q96J ST6G Beta-galactoside alpha-2,6- AHPAGSFHAGPGDLQK 886
 EFFSSQVGR 887
EGAFPAAQVQR 888
EVHVYEYIPSVR 889
IINSQILTNPSHHFIDSSLYK 890
LLPVQGK 891
LNMGTQGDLHR 892
LVPAVPLSQLHPR 893
NPNQPFYILHPK 894
RLLPVQGK 895
WAQSQDGFEHK 896
WAQSQDGFEHKEFFSSQVGR 897
Q9NT TENM Teneurin-2 AGHWFATTTPIIGK 898
 ALGTAWAK 899
ASGWSVQYR 900
DLAGNSEVVAGTGEQCLPFDEAR 901
FDYTYHDNSFR 902
FSEEGMVNAR 903
GIMFAIK 904
GSDIFEYNSK 905
HSMSTHTSIGYIR 906
IASIKPVISETPLPVDLYR 907
IDDDGYLCQR 908
IGSADGDLVTLGTTIGR 909
IKEVQYEMFR 910
ITENHQVSIIAGR 91 1
 KVASVLNNAYYLDK 912
LLAVTMPSVAR 913
MVNLQSGGFSCTIR 914
NVGKEPAPFNLYMFK 915
QYIFEYDSSDR 916
SDETGWTTFYDYDHEGR 917
SFQASPNLAYTFIWDK 918
SMVLLLQSQR 919
SNNPLSSELDLK 920
TNLGHHLQYFYSDLHNPTR 921
VASVLNNAYYLDK 922
VTTGVSSIASEDSR 923
VTTGVSSIASEDSRK 924
VWSYSYLDK 925
YGLTPDTLDEEK 926
YGYTITR 927
YSYDLNGNLHLLNPGNSVR 928
YTYDYDGDGQLQSVAVNDRPTWR 929
Q14D TME Transmembrane protein 132B EPGITTVQVLSPLSDSILAEK 930
 FSSLPAYLPTNLHISNAEESFFLK 931
GCSLQYQHATVR 932
IGSVVVYPTQDDLK 933
MFAFPEAR 934
SHILDSSIYSNRPK 935
TVIVLDDR 936
VEPFFIYR 937
WPIVVAEGEGQGPLIK 938
0951 UBL3 Ubiquitin-like protein 3 EFLFSPNDSASDIAK 939
ETLPEPNSQGQR 940
 FLHGNVTLGALK 941
LILVSGK 942
SSNVPADMINLR 943
TTVMHLVAR 944
P182 VCL Vinculin AGEVINQPMMMAAR 945
 ALASQLQDSLK 946
ALASQLQDSLKDLK 947
AQQVSQGLDVLTAK 948
AVAGNISDPGLQK 949
CDRVDQLTAQLADLAAR 950
DPSASPGDAGEQAIR 951
DYLIDGSR 952
ELLPVLISAMK 953
ELTPQVVSAAR 954
ETVQTTEDQILKR 955
EVENSEDPKFR 956
GNDIIAAAK 957
GQGSSPVAMQK 958
GWLRDPSASPGDAGEQAIR 959
IPTISTQLK 960
LANVMMGPYR 961
LLAVAATAPPDAPNREEVFDER 962
LVQAAQMLQSDPYSVPAR 963
MALLMAEMSR 964
MLGQMTDQVADLR 965
MSAEINEIIR 966
MTGLVDEAIDTK 967
NPGNQAAYEHFETMK 968
NQGIEEALK 969
NQWIDNVEK 970
QILDEAGK 971
QVATALQNLQTK 972
SFLDSGYR 973
SLGEISALTSK 974
SLLDASEEAIKK 975
STVEGIQASVK 976
TDAGFTLR 977
TNLLQVCER 978
VLQLTSWDEDAWASK 979
VMLVNSMNTVK 980
WIDNPTVDDR 981
References
[1] Alcolea D, Martinez-Lage P, Izagirre A, Clerigue M, Carmona- Iragui M, Alvarez RM, et al. Feasibility of Lumbar Puncture in the Study of
 Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: A Multicenter Study in Spain. J Alzheimers Dis 2013.
 [2] Arnold SE, Han LY, Clark CM, Grossman M, Trojanowski JQ.
Quantitative neurohistological features of frontotemporal degeneration. Neurobiol Aging 2000;21: 913-9.
 [3] Boddaert J, Kinugawa K, Lambert JC, Boukhtouche F, Zoll J,
Merval R, et al. Evidence of a role for lactadherin in Alzheimer's disease. Am J Pathol 2007; 170: 921-9.
 [4] Borroni B, Benussi A, Cosseddu M, Archetti S, Padovani A. Cerebrospinal Fluid Tau Levels Predict Prognosis in Non-Inherited Frontotemporal Dementia. Neurodegener Dis 2013.
 [5] Brettschneider J, Del TK, Irwin DJ, Grossman M, Robinson JL,
Toledo JB, et al. Sequential distribution of pTDP-43 pathology in behavioral variant frontotemporal dementia (bvFTD). Acta Neuropathol 2014.
 [6] Cairns NJ, Bigio EH, Mackenzie IR, Neumann M, Lee VM,
Hatanpaa KJ, et al. Neuropathologic diagnostic and nosologic criteria for
frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol 2007; 114: 5-22.
 [7] Cataltepe O, Arikan MC, Ghelfi E, Karaaslan C, Ozsurekci Y, Dresser K, et al. Fatty acid binding protein 4 is expressed in distinct endothelial and non-endothelial cell populations in glioblastoma. Neuropathol Appl Neurobiol 2012;38: 400- 10.
 [8] Comabella M, Fernandez M, Martin R, Rivera-Vallve S, Borras
E, Chiva C, et al. Cerebrospinal fluid chitinase 3-like 1 levels are associated with conversion to multiple sclerosis. Brain 2010; 133: 1082-93.
 [9] Craig- Schapiro R, Perrin RJ, Roe CM, Xiong C, Carter D, Cairns
NJ, et al. YKL-40: a novel prognostic fluid biomarker for preclinical Alzheimer's disease. Biol Psychiatry 2010;68: 903-12.  [10] Davidsson P, Sjogren M, Andreasen N, Lindbjer M, Nilsson CL, Westman-Brinkmalm A, et al. Studies of the pathophysiological mechanisms in frontotemporal dementia by proteome analysis of CSF proteins. Brain Res Mol Brain Res 2002; 109: 128-33.
 [11] Fratantoni SA, Piersma SR, Jimenez CR. Comparison of the performance of two affinity depletion spin filters for quantitative proteomics of CSF: Evaluation of sensitivity and reproducibility of CSF analysis using GeLC-MS/MS and spectral counting. Proteomics Clin Appl 2010;4: 613-7.
 [12] Hu WT, Watts K, Grossman M, Glass J, Lah JJ, Hales C, et al.
Reduced CSF p-Taul81 to Tau ratio is a biomarker for FTLD-TDP. Neurology 2013;81: 1945-52.
 [13] Huang dW, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 2009;37: 1-13.
 [14] Huang dW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009;4: 44-57.
 [15] Irwin DJ, Trojanowski JQ, Grossman M. Cerebrospinal fluid biomarkers for differentiation of frontotemporal lobar degeneration from Alzheimer's disease. Front Aging Neurosci 2013;5: 6.
 [16] Mattsson N, Ruetschi U, Pijnenburg YA, Blankenstein MA, Podust VN, Li S, et al. Novel cerebrospinal fluid biomarkers of axonal degeneration in frontotemporal dementia. Mol Med Rep 2008; 1: 757-61.
 [17] McMillan CT, Irwin DJ, Avants BB, Powers J, Cook PA, Toledo
JB, et al. White matter imaging helps dissociate tau from TDP-43 in frontotemporal lobar degeneration. J Neurol Neurosurg Psychiatry 2013;84: 949-55.
 [18] Neniskyte U, Brown GC. Lactadherin/MFG-E8 is essential for microglia-mediated neuronal loss and phagoptosis induced by amyloid beta. J Neurochem 2013; 126: 312-7.
 [19] Olsson B, Hertze J, Lautner R, Zetterberg H, Nagga K, Hoglund K, et al. Microglial markers are elevated in the prodromal phase of Alzheimer's disease and vascular dementia. J Alzheimers Dis 2013;33: 45-53.  [20] Pham TV, Piersma SR, Warmoes M, Jimenez CR. On the beta- binomial model for analysis of spectral count data in label-free tandem mass spectrometry-based proteomics. Bioinformatics 2010;26: 363-9.
 [21] Pijnenburg YA, Janssen JC, Schoonenboom NS, Petzold A, Mulder C, Stigbrand T, et al. CSF neurofilaments in frontotemporal dementia compared with early onset Alzheimer's disease and controls. Dement Geriatr Cogn Disord 2007;23: 225-30.
 [22] Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain 2011; 134: 2456-77.
 [23] Rentzos M, Paraskevas GP, Kapaki E, Nikolaou C, Zoga M, Rombos A, et al. Interleukin-12 is reduced in cerebrospinal fluid of patients with Alzheimer's disease and frontotemporal dementia. J Neurol Sci 2006;249: 110-4.
 [24] Ruetschi U, Zetterberg H, Podust VN, Gottfries J, Li S, Hviid SA, et al. Identification of CSF biomarkers for frontotemporal dementia using SELDI- TOF. Exp Neurol 2005; 196: 273-81.
 [25] Schofield EC, Halliday GM, Kwok J, Loy C, Double KL, Hodges JR. Low serum progranulin predicts the presence of mutations: a prospective study. J Alzheimers Dis 2010;22: 981-4.
 [26] Schoonenboom NS, Reesink FE, Verwey NA, Kester MI,
Teunissen CE, van de Ven PM, et al. Cerebrospinal fluid markers for differential dementia diagnosis in a large memory clinic cohort. Neurology 2012;78: 47-54.
 [27] Schweitzer K, Decker E, Zhu L, Miller RE, Mirra SS, Spina S, et al. Aberrantly regulated proteins in frontotemporal dementia. Biochem Biophys Res Commun 2006;348: 465-72.
 [28] Simonsen AH, McGuire J, Podust VN, Hagnelius NO, Nilsson TK, Kapaki E, et al. A novel panel of cerebrospinal fluid biomarkers for the
differential diagnosis of Alzheimer's disease versus normal aging and frontotemporal dementia. Dement Geriatr Cogn Disord 2007;24: 434-40.
 [29] Sjogren M, Folkesson S, Blennow K, Tarkowski E. Increased intrathecal inflammatory activity in frontotemporal dementia: pathophysiological implications. J Neurol Neurosurg Psychiatry 2004;75: 1107- 11.  [30] Sleegers K, Brouwers N, Van DP, Engelborghs S, Gijselinck I, van der Zee J, et al. Serum biomarker for progranulin-associated frontotemporal lobar degeneration. Ann Neurol 2009;65: 603-9.
 [31] Tso AW, Lam TK, Xu A, Yiu KH, Tse HF, Li LS, et al. Serum adipocyte fatty acid-binding protein associated with ischemic stroke and early death. Neurology 2011; 76: 1968-75.
 [32] von MC, Huynen M, Jaeggi D, Schmidt S, Bork P, Snel B.
STRING: a database of predicted functional associations between proteins. Nucleic Acids Res 2003;31: 258-61.
 [33] Working- Group. Consensus Report of the Working Group on:
"Molecular and Biochemical Markers of Alzheimer's Disease". The Ronald and Nancy Reagan Research Institute of the Alzheimer's Association and the National Institute on Aging Working Group. Neurobiol Aging 1998; 19: 109-16.