AU2012264688A2 - Methods for diagnosing multiple sclerosis - Google Patents

Methods for diagnosing multiple sclerosis Download PDF

Info

Publication number
AU2012264688A2
AU2012264688A2 AU2012264688A AU2012264688A AU2012264688A2 AU 2012264688 A2 AU2012264688 A2 AU 2012264688A2 AU 2012264688 A AU2012264688 A AU 2012264688A AU 2012264688 A AU2012264688 A AU 2012264688A AU 2012264688 A2 AU2012264688 A2 AU 2012264688A2
Authority
AU
Australia
Prior art keywords
biomarker
multiple sclerosis
subject
amount
ceramide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2012264688A
Other versions
AU2012264688A8 (en
AU2012264688A1 (en
Inventor
Susan Carvalho
Bernhard Hemmer
Ulrike Rennefahrt
Regina Reszka
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Metanomics Health GmbH
Original Assignee
Metanomics Health GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Metanomics Health GmbH filed Critical Metanomics Health GmbH
Publication of AU2012264688A1 publication Critical patent/AU2012264688A1/en
Publication of AU2012264688A8 publication Critical patent/AU2012264688A8/en
Publication of AU2012264688A2 publication Critical patent/AU2012264688A2/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/50Determining the risk of developing a disease

Abstract

The present invention relates to the field of diagnostic methods. Specifically, the present invention contemplates a method for diagnosing multiple sclerosis in a subject, a method for identifying whether a subject is in need for a therapy of multiple sclerosis or a method for determining whether a multiple sclerosis therapy is successful. Moreover, contributed is a method for diagnosing or predicting the risk of an active status of multiple sclerosis in a subject. The invention also relates to tools for carrying out the aforementioned methods, such as diagnostic devices.

Description

WO 2012/164026 PCT/EP2012/060262 - 1 METHODS FOR DIAGNOSING MULTIPLE SCLEROSIS The present invention relates to the field of diagnostic methods. Specifically, the present inven tion contemplates a method for diagnosing multiple sclerosis, a method for identifying whether a 5 subject is in need for a therapy of multiple sclerosis or a method for determining whether a mul tiple sclerosis therapy is successful. Moreover, contributed is a method for diagnosing or pre dicting the risk of an active status or the progression of multiple sclerosis in a subject. The in vention also relates to tools for carrying out the aforementioned methods, such as diagnostic devices. 10 Multiple sclerosis (MS) affects approximately 1 million individuals worldwide and is the most common disease of the central nervous system (CNS) that causes prolonged and severe disa bility in young adults. Although its etiology remains elusive, strong evidence supports the con cept that a T cell-mediated inflammatory process against self molecules within the white matter 15 of the brain and spinal cord underlies its pathogenesis. Since myelin-reactive T cells are present in both MS patients and healthy individuals, the primary immune abnormality in MS most likely involves failed regulatory mechanisms that lead to an enhanced T cell activation status and less stringent activation requirements. Thus, the pathogenesis includes activation of encephalitogen ic, i.e. autoimmune myelin-specific T cells outside the CNS, followed by: an opening of the 20 blood-brain barrier; T cell and macrophage infiltration; microglial activation; demyelination, and irreversible neuronal damage (Aktas 2005, Neuron 46, 421-432, Zamvil 2003, Neuron 38:685 688 or Zipp 2006, Trends Neurosci. 29, 518-527). While much is known about the mechanisms responsible for the encephalitogenicity of T cells, little is known as yet regarding the body's en dogenous control mechanisms for regulating harmful lymphocyte responses into and within the 25 CNS. In addition, despite extensive studies on T-cell mediated demyelination, the damage pro cesses in vivo within the CNS are not fully understood. Currently, diagnostic tools such as neuroimaging, analysis of cerebrospinal fluid and evoked potentials are used for diagnosing MS. Magnetic resonance imaging of the brain and spine can 30 visualize demyelination (lesions or plaques). Gadolinium can be administered intravenously as a contrast agent to mark active plaques and, by elimination, demonstrate the existence of his torical lesions which are not associated with symptoms at the moment of the evaluation. Analys ing cerebrospinal fluid obtained from a lumbar puncture can provide evidence of chronic in flammation of the central nervous system. The cerebrospinal fluid can be analyzed for oligoclo 35 nal bands, which are an inflammation marker found in 75-85% of people with MS (Link 2006, J Neuroimmunol. 180 (1-2): 17-28. However, none of the aforementioned techniques is specific to MS, only. Therefore, most often only biopsies or post-mortem examinations can yield a reliable diagnosis. 40 Multiple sclerosis is a chronic demyelinating disease of the central nervous system that pre sents with variable signs and symptoms. This variability in the clinical presentation may result in misdiagnosis, unnecessary referrals and misleading information to the patients. Since MS is a WO 2012/164026 PCT/EP2012/060262 -2 clinically highly heterogeneous disease, diagnostic and prognostic markers are needed to facili tate diagnose, predict the course of the disease in the individual patient, the necessity of treat ment and the kind of therapy (Bielekova & Martin 2004, Brain 127: 1463-1478). The response to the currently available therapies differs from patient to patient without any evidences from the 5 course of the disease. Markers would alleviate the choice of drug to apply, which will be even more important within the next years, when further drugs will come on the market. Furthermore, rapidly progressing patients should be treated from the beginning more aggressively than pa tients with a rather benign disease course. Markers of tissue damage and, in particular, neu ronal damage may be only or higher expressed in patients with rapid progression and subse 10 quent disability. On the other hand, treating the patients with an aggressive therapy with poten tially devastating side effects requires therapy response markers as well as a risk management. Thus biomarkers for disease activity and response to therapy are valuable for determining the patient's prognosis, and can allow a personalized adjustment of therapy. 15 Accordingly, means and methods for reliably diagnosing MS and for evaluating the success of a therapy are highly desired but not yet available. Therefore, the present invention relates to a method for diagnosing multiple sclerosis in a sub ject comprising the steps of: 2) determining in a sample of the subject the amount of at least one ceramide, or the total amount of ceramides, and b) comparing the amount of the said at least one ceramide, or the total amount of ceramides to a reference amount, whereby multiple sclerosis is to be diagnosed. 25 In particular, the present invention relates to a method for diagnosing multiple sclerosis in a sub ject comprising the steps of: a) determining in a sample of the subject the amount of at least one biomarker selected from the biomarkers listed in Table 1 and/or Table 2, and b) comparing the amount of the said at least one biomarker to a reference amount, whereby 30 multiple sclerosis is to be diagnosed. The method as referred to in accordance with the present invention includes a method which essentially consists of the aforementioned steps or a method which includes further steps. However, it is to be understood that the method, in a preferred embodiment, is a method carried 35 out ex vivo, i.e. not practised on the human or animal body. The method, preferably, can be assisted by automation. The term "diagnosing" as used herein refers to assessing whether a subject suffers from the disease MS, or not. As will be understood by those skilled in the art, such an assessment, alt 40 hough preferred to be, may usually not be correct for 100% of the investigated subjects. The term, however, requires that a statistically significant portion of subjects can be correctly as sessed and, thus, diagnosed. Whether a portion is statistically significant can be determined WO 2012/164026 PCT/EP2012/060262 -3 without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p-value determination, Student's t-test, Anno va, Mann-Whitney test, etc.. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 50%, at least 5 60%, at least 70%, at least 80%, at least 90%, at least 95%. The p-values are, preferably, 0.2, 0.1, 0.05, 0.01, 0.005, 0.001. The term includes individual diagnosis of MS or its symptoms as well as continuous monitoring of a patient. Monitoring, i.e. diagnosing the presence or absence of MS or the symptoms ac 10 companying it at various time points, includes monitoring of patients known to suffer from MS as well as monitoring of subjects known to be at risk of developing MS. Furthermore, monitoring can also be used to determine whether a patient is treated successfully or whether at least symptoms of MS can be ameliorated over time by a certain therapy. 15 The term "MS (multiple sclerosis)" as used herein relates to disease of the central nervous sys tem (CNS) that causes prolonged and severe disability in a subject suffering therefrom. How to diagnose MS is well known in the art. In particular, MS can be diagnosed according to the crite ria as disclosed in Polman et al. ("revised McDonald criteria", Ann Neurol 2005; 58:840-846) which herewith is incorporated by reference in its entirety with respect to its entire disclosure 20 content. The pathogenesis of MS includes activation of encephalitogenic, i.e. autoimmune mye lin-specific T cells outside the CNS, followed by an opening of the blood-brain barrier, T cell and macrophage infiltration, microglial activation, demyelination, and irreversible neuronal damage. There are four standardized subtype definitions of MS which are also encompassed by the term as used in accordance with the present invention: relapsing remitting, secondary progressive, 25 primary progressive and progressive relapsing. The relapsing-remitting subtype is characterized by unpredictable relapses followed by periods of months to years of remission with no new signs of disease activity. Deficits suffered during attacks (active status) may either resolve or leave sequelae. This describes the initial course of 85 to 90% of subjects suffering from MS. In cases of so-called benign MS the deficits always resolve between active status. Secondary pro 30 gressive MS describes those with initial relapsing-remitting MS, who then begin to have pro gressive neurological decline between acute attacks without any definite periods of remission. Occasional relapses and minor remissions may appear. The median time between disease on set and conversion from relapsing-remitting to secondary progressive MS is about 19 years. The primary progressive subtype describes about 10 to 15% of subjects who never have remis 35 sion after their initial MS symptoms. It is characterized by progression of disability from onset, with no, or only occasional and minor, remissions and improvements. The age of onset for the primary progressive subtype is later than other subtypes. Progressive relapsing MS describes those subjects who, from onset, have a steady neurological decline but also suffer clear super imposed attacks. This is the least common of all subtypes. There are also some cases of atypi 40 cal MS which can not be allocated in the aforementioned subtype groups. As used herein, the term "multiple sclerosis" also encompasses the clinically isolated syndrome (frequently also re ferred to as "CIS").
WO 2012/164026 PCT/EP2012/060262 -4 Symptoms associated with MS include changes in sensation (hypoesthesia and paraesthesia), muscle weakness, muscle spasms, difficulty in moving, difficulties with coordination and bal ance (ataxia), problems in speech (dysarthria) or swallowing (dysphagia), visual problems (nystagmus, optic neuritis, or diplopia), fatigue, headache, acute or chronic pain, bladder and 5 bowel difficulties. Cognitive impairment of varying degrees as well as emotional symptoms of depression or unstable mood may also occur as symptoms. The main clinical measure of disa bility progression and symptom severity is the Expanded Disability Status Scale (EDSS). Further symptoms of MS are well known in the art and are described in the standard text books 10 of medicine, such as Stedman or Pschyrembl. In the studies carried out in the context of the present invention, it was shown that the bi omarkers as shown in Table 1 and 2, are particularly useful for the early diagnosis of MS. Ac cordingly, the present invention particularly relates to a method for the early diagnosis of MS, 15 and, thus, to the diagnosis of MS in an early stage of MS. The early stage of MS encompasses the time period of, preferably, three years, more preferably, two years, even more preferably, one year, and most preferably, six months after the onset of MS. Also preferably, the early stage of MS encompasses the time period from the first acute neurological event (attack) of MS until the second acute neurological event (attack) of MS. It may further encompass the time pe 20 riod from the first acute neurological event (attack) of MS until the third acute neurological event (attack) of MS. A neurological event, preferably, refers to a demyelinating event, more prefera bly, to an inflammatory demyelinating event. A subject who is in an early stage of MS, prefera bly, has two lesions in the central nervous system. More preferably, said subject has one lesion in the central nervous system. A lesion, preferably, is an area of the central nervous system that 25 is inflamed and/or has been demyelinated. Such lesions can be, e.g., detected by MRI. Prefera bly, the subject who is in an early stage also shows symptoms of MS. Preferred symptoms, in particular neurological symptoms, are disclosed elsewhere herein. The term "biomarker" as used herein refers to a molecular species which serves as an indicator 30 for a disease or effect as referred to in this specification. Said molecular species can be a me tabolite itself which is found in a sample of a subject. Moreover, the biomarker may also be a molecular species which is derived from said metabolite. In such a case, the actual metabolite will be chemically modified in the sample or during the determination process and, as a result of said modification, a chemically different molecular species, i.e. the analyte, will be the deter 35 mined molecular species. It is to be understood that in such a case, the analyte represents the actual metabolite and has the same potential as an indicator for the respective medical condi tion. Moreover, a biomarker according to the present invention is not necessarily corresponding to 40 one molecular species. Rather, the biomarker may comprise stereoisomers or enantiomeres of a compound. Further, a biomarker can also represent the sum of isomers of a biological class of isomeric molecules. Said isomers shall exhibit identical analytical characteristics in some cases WO 2012/164026 PCT/EP2012/060262 -5 and are, therefore, not distinguishable by various analytical methods including those applied in the accompanying Examples described below. However, the isomers will share at least identical sum formula parameters and, thus, in the case of, e.g., lipids an identical chain length and iden tical numbers of double bonds in the fatty acid and/or sphingo base moieties. 5 The biomarker to be used in the context of the method of the present invention, is preferably, a ceramide. Ceramides are lipid molecules which are composed of aminoalcohol, coupled to a single fatty acid via amid linkage. Sphingosine (2-aminooctadec-4-ene-1,3-diol). Preferably, the amino alcohol is sphingosine (2-aminooctadec-4-ene-1,3-diol), a long chain aliphatic amine. 10 Other aminoalcohols, also referred to as "Long Chain Bases" (LCB), eg with different chain lengths such as d16-, d17- or d20-Sphingosine and also the respective saturated aminoalcohols (Dihydrosphingosine or Sphinganine) may occur. The nomenclature used to describe the differ ent Ceramide species is according to the "Lipidmaps" classification system as described in "Up date of the LIPID MAPS comprehensive classification system for lipids"; Fahy E., Subramaniam 15 S., Murphy R., Nishijima M., Raetz C., Shimizu T., Spener F., van Meer G., Wakelam M., and Dennis E., Journal of Lipid Research 50, S9-S14 (2009). The fatty acid coupled to sphingosine may have variable chain length and different degrees of saturation/unsaturation. Preferred ceramides are C21-, C22, C23 or C24 ceramides. Particularly 20 preferred ceramides to be used as a biomarker in the context of the method of the present in vention are listed herein below and/or in Table 1 and/or 2. Accordingly, the ceramide in the context of the methods of the present invention is preferably selected from the group of ceramides consisting of ceramide (d16:1,C22:0), ceramide 25 (dl6:1,C23:0), ceramide (d16:1,C24:0), ceramide (d17:1,C22:0), ceramide (d17:1,C23:0), ceramide (d17:1,C24:0), ceramide (d17:1,C24:1), ceramide (d18:1, C21:0), ceramide (d18:1,C22:0), ceramide (d18:1,C22:1), ceramide (d18:1, C23:0), ceramide (d18:1,C23:1), ceramide (d18:1, C24:0), ceramide (d18:1, C24:1), ceramide (d18:2,C22:0), ceramide (dl8:2,C23:0), ceramide (dl8:2,C24:0), and ceramide (dl8:2,C24:1). 30 In a further preferred embodiment, the ceramide is selected from the group of ceramides con sisting of ceramide (d16:1, C22:0), ceramide (d16:1, C23:0), ceramide (d16:1, C24:0), ceramide (d17:1, C22:0), ceramide (d17:1, C23:0), ceramide (d17:1, C24:0), ceramide (d17:1, C24:1), ceramide (d18:1, C21:0), ceramide (d18:1, C22:0), ceramide (d18:1, C22:1), ceramide (d18:1, 35 C23:0), ceramide (d18:1, C23:1), ceramide (d18:2,C22:0), ceramide (d18:2, C23:0), ceramide (d18:2, C24:0), and ceramide (d18:2,C24:1). In one embodiment, the ceramide is not ceramide (d18:1, C24:0) or not ceramide (d18:1, C24:1). Preferred methods for the determination of ceramides are disclosed in the Examples or, are 40 those known in the art, see, e.g. Kasumov et al., Analytical Biochemistry 401 (2010) 154 to 161 which hereby is incorporated by reference with respect to the entire disclosure content.
WO 2012/164026 PCT/EP2012/060262 -6 In the method according to the present invention, at least one metabolite of the aforementioned group of biomarkers, i.e. the biomarkers as shown in Table 1 and/or Table 2, is to be deter mined. However, more preferably, a group of biomarkers will be determined in order to strengthen specificity and/or sensitivity of the assessment. Such a group, preferably, comprises 5 at least 2, at least 3, at least 4, at least 5, at least 10 or up to all of the said biomarkers shown in the Tables. In addition to the specific biomarkers recited in the specification, other biomarkers may be, preferably, determined as well in the methods of the present invention. 10 In the context of the method of the present invention, it is also envisaged to determine in step a) of the methods of the present invention the total amount of ceramides in a sample instead of the amount of at least one biomarker as listed in Tables 1 to 3, in particular in Tables 1 and 2. Thus, the total ceramide content shall be determined. In step b) said total amount of ceramides shall be compared to a reference amount. Preferably, the total amount of C21 ceramides, C22 15 ceramides, C23 ceramides, or C24 ceramides is determined. More preferably, the total amount of C21 to C24 ceramides is determined. An increase of the total amount of ceramides as com pared to the reference is, preferably, indicative for the diagnosis of MS, in particularly for the diagnosis of an early stage of MS. 20 In a preferred embodiment of the method of the invention, said at least one biomarker is select ed from the group of biomarkers listed in Table 1 a and/or Table 2a. An increase in such a bi omarker is indicative for multiple sclerosis, in particular for an early stage of multiple sclerosis. Preferably, an increase of at least 10%, of at least 20%, of at least 30%, or of at least 50% is indicative for the diagnosis of MS (in an increasing order of preference). The same applies if the 25 biomarker is a ceramide. The same applies also if the total amount of ceramides is determined in the sample. In another preferred embodiment of the method of the present invention said at least one bi omarker is selected from the group of biomarkers listed in Table 1 b and/or Table 2b. A de 30 crease in such a biomarker is indicative for multiple sclerosis, in particular for an early stage of multiple sclerosis. Preferably, a decrease of at least 10%, of at least 20%, of at least 30%, of at least 50% is indicative for the diagnosis of MS (in an increasing order of preference). In a preferred embodiment of the method of the present invention, the amount of at least one 35 biomarker as shown in Table 2 is determined, if the subject to be tested is suspected to be in an acute stage of MS. A subject who suspected to be in an acute stage of MS, preferably, shows symptoms of the acute stage of MS, whereas a subject who is not suspected to be in an acute stage of MS, preferably, does not show symptoms of the acute stage of MS. 40 Preferably, the at least one biomarker is selected from the group of biomarkers listed in Table 2a if the subject to be tested is suspected to be in an acute stage of MS. An increase in such a WO 2012/164026 PCT/EP2012/060262 -7 biomarker is indicative for multiple sclerosis. Said increase is preferably an increase of at least 10%, of at least 20%, of at least 30%, of at least 50% (in an increasing order of preference). 5 Preferably, the at least one biomarker is selected from the group of biomarkers listed in Table 2b if the subject to be tested is suspected to be in an acute stage of MS. A decrease in such a biomarker is indicative for multiple sclerosis. A metabolite as used herein refers to at least one molecule of a specific metabolite up to a plu 10 rality of molecules of the said specific metabolite. It is to be understood further that a group of metabolites means a plurality of chemically different molecules wherein for each metabolite at least one molecule up to a plurality of molecules may be present. A metabolite in accordance with the present invention encompasses all classes of organic or inorganic chemical com pounds including those being comprised by biological material such as organisms. Preferably, 15 the metabolite in accordance with the present invention is a small molecule compound. More preferably, in case a plurality of metabolites is envisaged, said plurality of metabolites repre senting a metabolome, i.e. the collection of metabolites being comprised by an organism, an organ, a tissue, a body fluid or a cell at a specific time and under specific conditions. 20 The metabolites are small molecule compounds, such as substrates for enzymes of metabolic pathways, intermediates of such pathways or the products obtained by a metabolic pathway. Metabolic pathways are well known in the art and may vary between species. Preferably, said pathways include at least citric acid cycle, respiratory chain, glycolysis, gluconeogenesis, hex ose monophosphate pathway, oxidative pentose phosphate pathway, production and P 25 oxidation of fatty acids, urea cycle, amino acid biosynthesis pathways, protein degradation pathways such as proteasomal degradation, amino acid degrading pathways, biosynthesis or degradation of: lipids, polyketides (including e.g. flavonoids and isoflavonoids), isoprenoids (in cluding eg. terpenes, sterols, steroids, carotenoids, xanthophylls), carbohydrates, phenylpro panoids and derivatives, alcaloids, benzenoids, indoles, indole-sulfur compounds, porphyrines, 30 anthocyans, hormones, vitamins, cofactors such as prosthetic groups or electron carriers, lignin, glucosinolates, purines, pyrimidines, nucleosides, nucleotides and related molecules such as tRNAs, microRNAs (miRNA) or mRNAs. Accordingly, small molecule compound metabolites are preferably composed of the following classes of compounds: alcohols, alkanes, alkenes, alkines, aromatic compounds, ketones, aldehydes, carboxylic acids, esters, amines, imines, 35 amides, cyanides, amino acids, peptides, thiols, thioesters, phosphate esters, sulfate esters, thioethers, sulfoxides, ethers, or combinations or derivatives of the aforementioned compounds. The small molecules among the metabolites may be primary metabolites which are required for normal cellular function, organ function or animal growth, development or health. Moreover, small molecule metabolites further comprise secondary metabolites having essential ecological 40 function, e.g. metabolites which allow an organism to adapt to its environment. Furthermore, metabolites are not limited to said primary and secondary metabolites and further encompass artificial small molecule compounds. Said artificial small molecule compounds are derived from WO 2012/164026 PCT/EP2012/060262 -8 exogenously provided small molecules which are administered or taken up by an organism but are not primary or secondary metabolites as defined above. For instance, artificial small mole cule compounds may be metabolic products obtained from drugs by metabolic pathways of the animal. Moreover, metabolites further include peptides, oligopeptides, polypeptides, oligonucle 5 otides and polynucleotides, such as RNA or DNA. More preferably, a metabolite has a molecu lar weight of 50 Da (Dalton) to 30,000 Da, most preferably less than 30,000 Da, less than 20,000 Da, less than 15,000 Da, less than 10,000 Da, less than 8,000 Da, less than 7,000 Da, less than 6,000 Da, less than 5,000 Da, less than 4,000 Da, less than 3,000 Da, less than 2,000 Da, less than 1,000 Da, less than 500 Da, less than 300 Da, less than 200 Da, less than 100 10 Da. Preferably, a metabolite has, however, a molecular weight of at least 50 Da. Most prefera bly, a metabolite in accordance with the present invention has a molecular weight of 50 Da up to 1,500 Da. The term "sample" as used herein refers to samples from body fluids, preferably, blood, plasma, 15 serum, saliva, urine or cerebrospinal fluid, or samples derived, e.g., by biopsy, from cells, tis sues or organs, in particular from the CNS including brain and spine. More preferably, the sam ple is a blood, plasma or serum sample, most preferably, a serum or plasma sample. Biological samples can be derived from a subject as specified elsewhere herein. Techniques for obtaining the aforementioned different types of biological samples are well known in the art. For example, 20 blood samples may be obtained by blood taking while tissue or organ samples are to be ob tained, e.g., by biopsy. The aforementioned samples are, preferably, pre-treated before they are used for the method of the present invention. As described in more detail below, said pre-treatment may include treat 25 ments required to release or separate the compounds or to remove excessive material or waste. Suitable techniques comprise centrifugation, extraction, fractioning, ultrafiltration, protein precipitation followed by filtration and purification and/or enrichment of compounds. Moreover, other pre-treatments are carried out in order to provide the compounds in a form or concentra tion suitable for compound analysis. For example, if gas-chromatography coupled mass spec 30 trometry is used in the method of the present invention, it will be required to derivatize the com pounds prior to the said gas chromatography. Suitable and necessary pre-treatments depend on the means used for carrying out the method of the invention and are well known to the per son skilled in the art. Pre-treated samples as described before are also comprised by the term "sample" as used in accordance with the present invention. 35 The term "subject" as used herein relates to animals and, preferably, to mammals. More prefer ably, the subject is a primate and, most preferably, a human. The subject, preferably, is sus pected to suffer from MS, i.e. it may already show some or all of the symptoms associated with the disease. 40 The term "determining the amount" as used herein refers to determining at least one character istic feature of a biomarker to be determined by the method of the present invention in the sam- WO 2012/164026 PCT/EP2012/060262 -9 ple. Characteristic features in accordance with the present invention are features which charac terize the physical and/or chemical properties including biochemical properties of a biomarker. Such properties include, e.g., molecular weight, viscosity, density, electrical charge, spin, optical activity, colour, fluorescence, chemoluminescence, elementary composition, chemical structure, 5 capability to react with other compounds, capability to elicit a response in a biological read out system (e.g., induction of a reporter gene) and the like. Values for said properties may serve as characteristic features and can be determined by techniques well known in the art. Moreover, the characteristic feature may be any feature which is derived from the values of the physical and/or chemical properties of a biomarker by standard operations, e.g., mathematical calcula 10 tions such as multiplication, division or logarithmic calculus. Most preferably, the at least one characteristic feature allows the determination and/or chemical identification of the said at least one biomarker and its amount. Accordingly, the characteristic value, preferably, also comprises information relating to the abundance of the biomarker from which the characteristic value is derived. For example, a characteristic value of a biomarker may be a peak in a mass spectrum. 15 Such a peak contains characteristic information of the biomarker, i.e. the m/z information (mass/charge ratio or quotient), as well as an intensity value being related to the abundance of the said biomarker (i.e. its amount) in the sample. As discussed before, each biomarker comprised by a sample may be, preferably, determined in 20 accordance with the present invention quantitatively or semi-quantitatively. For quantitative de termination, either the absolute or precise amount of the biomarker will be determined or the relative amount of the biomarker will be determined based on the value determined for the characteristic feature(s) referred to herein above. The relative amount may be determined in a case were the precise amount of a biomarker can or shall not be determined. In said case, it 25 can be determined whether the amount in which the biomarker is present is enlarged or dimin ished with respect to a second sample comprising said biomarker in a second amount. In a pre ferred embodiment said second sample comprising said biomarker shall be a calculated refer ence as specified elsewhere herein. Quantitatively analysing a biomarker, thus, also includes what is sometimes referred to as semi-quantitative analysis of a biomarker. 30 Moreover, determining as used in the method of the present invention, preferably, includes us ing a compound separation step prior to the analysis step referred to before. Preferably, said compound separation step yields a time resolved separation of the metabolites comprised by the sample. Suitable techniques for separation to be used preferably in accordance with the 35 present invention, therefore, include all chromatographic separation techniques such as liquid chromatography (LC), high performance liquid chromatography (HPLC), gas chromatography (GC), thin layer chromatography, size exclusion or affinity chromatography. These techniques are well known in the art and can be applied by the person skilled in the art without further ado. Most preferably, LC and/or GC are chromatographic techniques to be envisaged by the method 40 of the present invention. Suitable devices for such determination of biomarkers are well known in the art. Preferably, mass spectrometry is used in particular gas chromatography mass spec trometry (GC-MS), liquid chromatography mass spectrometry (LC-MS), direct infusion mass WO 2012/164026 PCT/EP2012/060262 -10 spectrometry or Fourier transform ion-cyclotrone-resonance mass spectrometry (FT-ICR-MS), capillary electrophoresis mass spectrometry (CE-MS), high-performance liquid chromatography coupled mass spectrometry (HPLC-MS), quadrupole mass spectrometry, any sequentially cou pled mass spectrometry, such as MS-MS or MS-MS-MS, inductively coupled plasma mass 5 spectrometry (ICP-MS), pyrolysis mass spectrometry (Py-MS), ion mobility mass spectrometry or time of flight mass spectrometry (TOF). Most preferably, LC-MS and/or GC-MS are used as described in detail below. Said techniques are disclosed in, e.g., Nissen 1995, Journal of Chro matography A, 703: 37-57, US 4,540,884 or US 5,397,894, the disclosure content of which is hereby incorporated by reference. As an alternative or in addition to mass spectrometry tech 10 niques, the following techniques may be used for compound determination: nuclear magnetic resonance (NMR), magnetic resonance imaging (MRI), Fourier transform infrared analysis (FT IR), ultraviolet (UV) spectroscopy, refraction index (RI), fluorescent detection, radiochemical detection, electrochemical detection, light scattering (LS), dispersive Raman spectroscopy or flame ionisation detection (FID). These techniques are well known to the person skilled in the 15 art and can be applied without further ado. The method of the present invention shall be, prefer ably, assisted by automation. For example, sample processing or pre-treatment can be auto mated by robotics. Data processing and comparison is, preferably, assisted by suitable comput er programs and databases. Automation as described herein before allows using the method of the present invention in high-throughput approaches. 20 Moreover, the at least one biomarker can also be determined by a specific chemical or biologi cal assay. Said assay shall comprise means which allow to specifically detect the at least one biomarker in the sample. Preferably, said means are capable of specifically recognizing the chemical structure of the biomarker or are capable of specifically identifying the biomarker 25 based on its capability to react with other compounds or its capability to elicit a response in a biological read out system (e.g., induction of a reporter gene). Means which are capable of specifically recognizing the chemical structure of a biomarker are, preferably, antibodies or oth er proteins which specifically interact with chemical structures, such as receptors or enzymes. Specific antibodies, for instance, may be obtained using the biomarker as antigen by methods 30 well known in the art. Antibodies as referred to herein include both polyclonal and monoclonal antibodies, as well as fragments thereof, such as Fv, Fab and F(ab) 2 fragments that are capable of binding the antigen or hapten. The present invention also includes humanized hybrid antibod ies wherein amino acid sequences of a non-human donor antibody exhibiting a desired antigen specificity are combined with sequences of a human acceptor antibody. Moreover, encom 35 passed are single chain antibodies. The donor sequences will usually include at least the anti gen-binding amino acid residues of the donor but may comprise other structurally and/or func tionally relevant amino acid residues of the donor antibody as well. Such hybrids can be pre pared by several methods well known in the art. Suitable proteins which are capable of specifi cally recognizing the biomarker are, preferably, enzymes which are involved in the metabolic 40 conversion of the said biomarker. Said enzymes may either use the biomarker as a substrate or may convert a substrate into the biomarker. Moreover, said antibodies may be used as a basis to generate oligopeptides which specifically recognize the biomarker. These oligopeptides shall, WO 2012/164026 PCT/EP2012/060262 - 11 for example, comprise the enzyme's binding domains or pockets for the said biomarker. Suita ble antibody and/or enzyme based assays may be RIA (radioimmunoassay), ELISA (enzyme linked immunosorbent assay), sandwich enzyme immune tests, electrochemiluminescence sandwich immunoassays (ECLIA), dissociation-enhanced lanthanide fluoro immuno assay 5 (DELFIA) or solid phase immune tests. Moreover, the biomarker may also be determined based on its capability to react with other compounds, i.e. by a specific chemical reaction. Further, the biomarker may be determined in a sample due to its capability to elicit a response in a biological read out system. The biological response shall be detected as read out indicating the presence and/or the amount of the biomarker comprised by the sample. The biological response may be, 10 e.g., the induction of gene expression or a phenotypic response of a cell or an organism. In a preferred embodiment the determination of the least one biomarker is a quantitative process, e.g., allowing also the determination of the amount of the at least one biomarker in the sample As described above, said determining of the at least one biomarker can, preferably, comprise 15 mass spectrometry (MS). Mass spectrometry as used herein encompasses all techniques which allow for the determination of the molecular weight (i.e. the mass) or a mass variable corre sponding to a compound, i.e. a biomarker, to be determined in accordance with the present in vention. Preferably, mass spectrometry as used herein relates to GC-MS, LC-MS, direct infu sion mass spectrometry, FT-ICR-MS, CE-MS, HPLC-MS, quadrupole mass spectrometry, any 20 sequentially coupled mass spectrometry such as MS-MS or MS-MS-MS, ICP-MS, Py-MS, TOF or any combined approaches using the aforementioned techniques. How to apply these tech niques is well known to the person skilled in the art. Moreover, suitable devices are commercial ly available. More preferably, mass spectrometry as used herein relates to LC-MS and/or GC MS, i.e. to mass spectrometry being operatively linked to a prior chromatographic separation 25 step. More preferably, mass spectrometry as used herein encompasses quadrupole MS. Most preferably, said quadrupole MS is carried out as follows: a) selection of a mass/charge quotient (m/z) of an ion created by ionisation in a first analytical quadrupole of the mass spectrometer, b) fragmentation of the ion selected in step a) by applying an acceleration voltage in an additional subsequent quadrupole which is filled with a collision gas and acts as a collision chamber, c) 30 selection of a mass/charge quotient of an ion created by the fragmentation process in step b) in an additional subsequent quadrupole, whereby steps a) to c) of the method are carried out at least once and analysis of the mass/charge quotient of all the ions present in the mixture of substances as a result of the ionisation process, whereby the quadrupole is filled with collision gas but no acceleration voltage is applied during the analysis. Details on said most preferred 35 mass spectrometry to be used in accordance with the present invention can be found in WO 03/073464. More preferably, said mass spectrometry is liquid chromatography (LC) MS and/or gas chroma tography (GC) MS. Liquid chromatography as used herein refers to all techniques which allow 40 for separation of compounds (i.e. metabolites) in liquid or supercritical phase. Liquid chromatog raphy is characterized in that compounds in a mobile phase are passed through the stationary phase. When compounds pass through the stationary phase at different rates they become WO 2012/164026 PCT/EP2012/060262 - 12 separated in time since each individual compound has its specific retention time (i.e. the time which is required by the compound to pass through the system). Liquid chromatography as used herein also includes HPLC. Devices for liquid chromatography are commercially available, e.g. from Agilent Technologies, USA. Gas chromatography as applied in accordance with the 5 present invention, in principle, operates comparable to liquid chromatography. However, rather than having the compounds (i.e. metabolites) in a liquid mobile phase which is passed through the stationary phase, the compounds will be present in a gaseous volume. The compounds pass the column which may contain solid support materials as stationary phase or the walls of which may serve as or are coated with the stationary phase. Again, each compound has a spe 10 cific time which is required for passing through the column. Moreover, in the case of gas chro matography it is preferably envisaged that the compounds are derivatised prior to gas chroma tography. Suitable techniques for derivatisation are well known in the art. Preferably, derivatisa tion in accordance with the present invention relates to methoxymation and trimethylsilylation of, preferably, polar compounds and transmethylation, methoxymation and trimethylsilylation of, 15 preferably, non-polar (i.e. lipophilic) compounds. The term "reference" refers to values of characteristic features of each of the biomarker which can be correlated to a medical condition, i.e. the presence or absence of the disease, diseases status or an effect referred to herein. If the biomarker is selected from the biomarkers shown in 20 Table 1 a, 2a, or 3a, a reference is, preferably, a threshold amount for a biomarker whereby amounts found in a sample to be investigated which are higher than or essentially identical to the threshold are indicative for the presence of a medical condition while those being lower are indicative for the absence of the medical condition. If the biomarker is selected from the bi omarkers shown in Table 1 b or 2b, or 3b, a reference is, preferably a threshold amount for a 25 biomarker whereby amounts found in a sample to be investigated which are lower or identical than the threshold are indicative for the presence of a medical condition while those being high er are indicative for the absence of the medical condition. In accordance with the aforementioned method of the present invention, a reference is, prefera 30 bly, a reference amount obtained from a sample from a subject known to suffer from MS. In par ticular, the reference amount is obtained from a sample from a subject known to be in an early stage of MS. Preferably, an amount for the at least one biomarker found in the test sample be ing essentially identical is indicative for the presence of the disease. 35 Moreover, the reference, also preferably, could be from a subject known not to suffer from MS, preferably, an apparently healthy subject. More preferably, the reference could be derived from a subject known not to suffer from MS, but showing symptoms of MS (or from a group of said subjects). Accordingly, said subject shall show MS-like symptoms. The MS or MS-like symp toms of said subject shall be caused by neurological diseases other than MS. Preferred symp 40 toms are disclosed elsewhere herein. Moreover, said symptoms shall be preferentially symp toms similar or identical to MS symptoms and include changes in sensation (hypoesthesia and paraesthesia), muscle weakness, muscle spasms, difficulty in moving, difficulties with coordina- WO 2012/164026 PCT/EP2012/060262 -13 tion and balance (ataxia), problems in speech (dysarthria) or swallowing (dysphagia), visual problems (nystagmus, optic neuritis, or diplopia), fatigue, headache. If the reference amount is derived from the aforementioned subject, or group thereof, an amount for the at least one bi omarker found in the test sample being altered with respect to the reference is indicative for the 5 presence of the disease. Preferably, said amount is being altered by at least 10%, by at least 20%, by at least 30%, by at least 50%. Preferably, an amount of the biomarker in the test sam ple being increased as compared to the reference is indicative for the presence of MS, if the biomarker is selected from the group of biomarkers shown in Table 1 a or 2a (and if the bi omarker is a ceramide, or if the total amount of ceramides is determined). Preferably, an 10 amount of the biomarker in the test sample being decreased as compared to the reference is indicative for the presence of MS, if the biomarker is selected from the group of biomarkers shown in Table 1b or 2b. The same applies mutatis mutandis for a calculated reference, most preferably the average or 15 median, for the relative or absolute amount of the at least one biomarker of a population of indi viduals comprising the subject to be investigated. The absolute or relative amounts of the at least one biomarker of said individuals of the population can be determined as specified else where herein. How to calculate a suitable reference value, preferably, the average or median, is well known in the art. The population of subjects referred to before shall comprise a plurality of 20 subjects, preferably, at least 5, 10, 50, 100, 1,000 or 10,000 subjects. It is to be understood that the subject to be diagnosed by the method of the present invention and the subjects of the said plurality of subjects are of the same species. The amounts of the test sample and the reference amounts are essentially identical, if the val 25 ues for the characteristic features and, in the case of quantitative determination, the intensity values are essentially identical. Essentially identical means that the difference between two amounts is, preferably, not significant and shall be characterized in that the values for the inten sity are within at least the interval between 1st and 99th percentile, 5 th and 9 5 th percentile, 1 0th and 90th percentile, 20th and 80th percentile, 30th and 70th percentile, 40th and 60th percentile of 30 the reference value, preferably, the 50th, 60th, 70th, 80th, 90th or 9 5 th percentile of the reference value. Statistical test for determining whether two amounts are essentially identical are well known in the art and are also described elsewhere herein. An observed difference for two amounts, on the other hand, shall be statistically significant. A 35 difference in the relative or absolute amount is, preferably, significant outside of the interval be tween 4 5 th and 5 5 th percentile, 40th and 60th percentile, 30th and 70th percentile, 20th and 80th percentile, 10th and 90th percentile, 5 th and 9 5 th percentile, 1st and 99th percentile of the reference value. Preferred changes and fold-regulations are described in the accompanying Tables as well as in the Examples. 40 WO 2012/164026 PCT/EP2012/060262 - 14 Preferably, the reference, i.e. values for at least one characteristic features of the at least one biomarker, will be stored in a suitable data storage medium such as a database and are, thus, also available for future assessments. 5 The term "comparing" refers to determining whether the determined amount of a biomarker is essentially identical to a reference or differs therefrom. Preferably, a biomarker is deemed to differ from a reference if the observed difference is statistically significant which can be deter mined by statistical techniques referred to elsewhere in this description. If the difference is not statistically significant, the biomarker amount and the reference amount are essentially identical. 10 Based on the comparison referred to above, a subject can be assessed to suffer from the dis ease, or not. For the specific biomarkers referred to in this specification, preferred values for the changes in the relative amounts (i.e. "fold"- regulation) or the kind of regulation (i.e. "up"- or "down" 15 regulation resulting in a higher or lower relative and/or absolute amount) are indicated in the following Tables and in the Examples below. If it is indicated in said Table that a given bi omarker is "up- regulated" in a subject, the relative and/or absolute amount will be increased, if it is "down-regulated", the relative and/or absolute amount of the biomarker will be decreased. In the case of "up"-regulation the ratio of median shall exceed 1.0 while it will be below 1.0 in the 20 case of a "down"-regulation. Moreover, the "fold"-change indicates the degree of increase or decrease, e.g., a 2-fold increase means that the median of one group, e.g., the MS group, is twice the median of the biomarker of the other group, e.g., the reference group. Accordingly, the direction of regulation can be derived from the Tables as well. 25 The comparison is, preferably, assisted by automation. For example, a suitable computer pro gram comprising algorithms for the comparison of two different data sets (e.g., data sets com prising the values of the characteristic feature(s)) may be used. Such computer programs and algorithm are well known in the art. Notwithstanding the above, a comparison can also be car ried out manually. 30 Advantageously, it has been found in the study underlying the present invention that the amounts of the specific biomarkers referred to above are indicators for MS. Accordingly, the at least one biomarker as specified above in a sample can, in principle, be used for assessing whether a subject suffers from MS. This is particularly helpful for an efficient diagnosis of the 35 disease as well as for improving of the pre-clinical and clinical management of MS as well as an efficient monitoring of patients. Moreover, the findings underlying the present invention will also facilitate the development of efficient drug-based therapies against MS as set forth in detail be low. 40 The definitions and explanations of the terms made above apply mutatis mutandis for the follow ing embodiments of the present invention except specified otherwise herein below.
WO 2012/164026 PCT/EP2012/060262 -15 Moreover, the present invention relates to a method for differentiating between i) a subject showing symptoms of multiple sclerosis and suffering from multiple sclerosis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclerosis, said method comprising the steps of a determining in a sample of a subject showing symptoms of multiple sclerosis the amount of at least one ceramide, or the total amount of ceramides, and b) comparing the amount of the said at least one ceramide, or the total amount of ceramides, to at least one reference amount, whereby it is differentiated between i) and ii). 10 In particular, the present invention relates to a method for differentiating between i) a subject showing symptoms of multiple sclerosis and suffering from multiple sclerosis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclerosis, said method comprising the steps of a) determining in a sample of a subject showing symptoms of multiple sclerosis the amount of at 15 least one biomarker selected from the biomarkers listed in Table 1 and/or Table 2, and b) comparing the amount of the said at least one biomarker to at least one reference amount, whereby it is differentiated between i) and ii). The term "differentiating" as used herein, preferably, means to distinguish between a subject 20 showing symptoms of multiple sclerosis and suffering from multiple sclerosis and a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclerosis. Thus, the aforementioned method allows for assessing whether a subject who shows symptoms of multi ple sclerosis suffers from MS or not. As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for 100% of the subjects to be differentially 25 diagnosed. The term, however, requires that a statistically significant portion of subjects can be correctly diagnosed. Whether a diagnosis is correct can be confirmed by methods well known in the art. Moreover, whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., de termination of confidence intervals, p- value determination, Student's t-test, Mann- Whitney test, 30 ANOVA etc.. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99 %. The p-values are, preferably, 0.2, 0.1, 0.05, 0.01, 0.005, or 0.0001. 35 In a preferred embodiment of the aforementioned method, it is differentiated between i) a sub ject showing symptoms of multiple sclerosis and suffering from an early stage of multiple sclero sis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple scle rosis. 40 The term "subject" has been defined elsewhere herein. In accordance with the aforementioned method the subject, preferably, shall show symptoms of MS, in particular neurological symp toms of MS. A subject who shows symptoms of MS, but does not suffer from MS, preferably, WO 2012/164026 PCT/EP2012/060262 -16 suffers from a neurological disease other than MS. Preferred diseases are disclosed in the Ex amples section. Symptoms of MS have been described elsewhere herein. Preferred symptoms include changes 5 in sensation (hypoesthesia and paraesthesia), muscle weakness, muscle spasms, difficulty in moving, difficulties with coordination and balance (ataxia), problems in speech (dysarthria) or swallowing (dysphagia), visual problems (nystagmus, optic neuritis, or diplopia), fatigue, head ache. Preferably, a subject showing symptoms of MS shall suffer from headache, in particular migraine, hypaesthesia, paresthesia, or meningitis or from combinations thereof. 10 In a preferred embodiment of the method of the invention, said at least one biomarker is select ed from the group of biomarkers listed in Table 1 a and/or Table 2a. An increase in such a bi omarker as compared to the reference, preferably, indicates that the subject showing symptoms of MS suffers from MS, whereas an unchanged amount or a decrease of such as biomarker as 15 compared to the reference, preferably, indicates that the subject showing symptoms of MS does not suffer from MS. The same applies if the biomarker is a ceramide. The same also applies if the total amount of ceramides is determined in the sample. Preferred references have been disclosed elsewhere herein. 20 Preferably, the aforementioned increase or decrease is an increase/decrease of at least 10%, of at least 20%, of at least 30%, or of at least 50% (in an increasing order of preference). In a further preferred embodiment of the method of the invention, said at least one biomarker is selected from the group of biomarkers listed in Table 1b and/or Table 2b. A decrease in such a 25 biomarker as compared to the reference, preferably, indicates that the subject showing symp toms of MS suffers from MS, whereas an and unchanged amount or increase of in such a bi omarker as compared to the reference, preferably, indicates that the subject showing symptoms of MS does not suffer from MS. Preferred references have been disclosed above. 30 Preferably, the at least one biomarker is selected from the group of biomarkers listed in Table 2, 2a and 2b, respectively, if the subject to be tested is suspected to be in an acute stage of MS. The present invention also relates to a method for identifying whether a subject is in need for a therapy of multiple sclerosis comprising the steps of the aforementioned method of diagnosing 35 MS and the further step of identifying a subject in need if multiple sclerosis is diagnosed. The phrase "in need for a therapy of multiple sclerosis" as used herein means that the disease in the subject is in a status where therapeutic intervention is necessary or beneficial in order to ameliorate or treat MS or the symptoms associated therewith. Accordingly, the findings of the 40 studies underlying the present invention do not only allow diagnosing MS in a subject but also allow for identifying subjects which should be treated by an MS therapy. Once the subject has WO 2012/164026 PCT/EP2012/060262 -17 been identified, the method may further include a step of making recommendations for a thera py of MS. A therapy of multiple sclerosis as used in accordance with the present invention, preferably, 5 relates to a therapy which comprises or consists of the administration of at least one drug se lected from the group consisting of: Interferon Betala, Interferon Beta 1b, Azathioprin, Cyclo phosphamide, Glatiramer Acetate, Immunglobuline Methotrexat, Mitoxantrone, Leustatin, IVIg, Natalizumab, Teriflunomid, Statins, Daclizumab, Alemtuzumab, Ritximab, Sphingosin 1 phos phate antagonist Fingolimod (FTY720), Cladribine, Fumarate, Laquinimod, drugs affecting B 10 cells, and antisense agents and agents against CD49d. Also, the present invention contemplates a method for determining whether a multiple sclerosis therapy is successful comprising the steps of: a) determining at least one ceramide, or the total amount of ceramides, in a first and a second 15 sample of the subject wherein said first sample has been taken prior to or at the onset of the multiple sclerosis therapy and said second sample has been taken after the onset of the said therapy; and b) comparing the amount of the said at least one ceramide, or the total amount of ceramides, in the first sample to the amount or total amount in the second sample. 20 In the context of the aforementioned method, a decrease in the amount determined in the se cond sample in comparison to the first sample is, preferably, indicative for multiple sclerosis therapy being successful. An increase in the amount determined in the second sample in com parison to the first sample, preferably, indicates that multiple sclerosis therapy is not successful. 25 Said increase is preferably an increase of at least 10%, of at least 20%, of at least 30%, or of at least 50% (in an increasing order of preference). The term "ceramide" has been described elsewhere herein. Preferably, the ceramide is selected from the ceramides listed in Table 1 and/or Table 2. 30 Accordingly, the ceramide in the context of the methods of the present invention is preferably selected from the group of ceramides consisting of ceramide (d16:1,C22:0), ceramide (d16:1,C23:0), ceramide (d16:1,C24:0), ceramide (d17:1,C22:0), ceramide (d17:1,C23:0), ceramide (d17:1,C24:0), ceramide (d17:1,C24:1), ceramide (d18:1,C21:0), ceramide 35 (d18:1,C22:0), ceramide (d18:1,C22:1), ceramide (d18:1,C23:0), ceramide (d18:1,C23:1), ceramide (d18:1, C24:0), ceramide (d18:1, C24:1), ceramide (d18:2,C22:0), ceramide (d18:2,C23:0), ceramide (d18:2,C24:0), and ceramide (d18:2,C24:1). In a further preferred embodiment, the ceramide is selected from the group of ceramides con 40 sisting of ceramide (d16:1, C22:0), ceramide (d16:1, C23:0), ceramide (d16:1, C24:0), ceramide (d17:1, C22:0), ceramide (d17:1, C23:0), ceramide (d17:1, C24:0), ceramide (d17:1, C24:1), ceramide (d18:1, C21:0), ceramide (d18:1, C22:0), ceramide (d18:1, C22:1), ceramide (d18:1, WO 2012/164026 PCT/EP2012/060262 -18 C23:0), ceramide (d18:1, C23:1), ceramide (d18:2,C22:0), ceramide (d18:2, C23:0), ceramide (d18:2, C24:0), and ceramide (d18:2,C24:1). In one embodiment, the ceramide is not ceramide (d18:1, C24:0) or not ceramide (d18:1, C24:1). 5 Moreover, the present invention contemplates a method for determining whether a multiple scle rosis therapy is successful comprising the steps of: a) determining at least one biomarker selected from the biomarkers listed in Table 1 and/or Table 2 in a first and a second sample of the subject wherein said first sample has been taken prior to or at the onset of the multiple sclerosis therapy and said second sample has been taken after 10 the onset of the said therapy; and b) comparing the amount of the said at least one biomarker in the first sample to the amount in the second sample, whereby a change in the amount determined in the second sample in compari son to the first sample is indicative for multiple sclerosis therapy being successful. 15 It is to be understood that an MS therapy will be successful if MS or at least some symptoms thereof can be treated or ameliorated compared to an untreated subject. Moreover, a therapy is also successful as meant herein if the disease progression can be prevented or at least slowed down compared to an untreated subject. 20 In a preferred embodiment of the aforementioned methods, the at least one biomarker is select ed from the biomarkers listed in Table 1a and/or 2a. In such a case, the change in step b) is a decrease. Accordingly, a decrease, preferably, indicates that the multiple sclerosis therapy is successful. Said decrease is preferably a decrease of at least 10%, of at least 20%, of at least 30%, of at least 50% (in an increasing order of preference). The same applies if the biomarker 25 is a ceramide. The same also applies if the total amount of ceramides is determined in the sam ple. In yet another preferred embodiment of the aforementioned methods, the at least one biomarker is selected from the biomarkers listed in Table 1b and/or 2b. In such a case, the change in step 30 b) is an increase. Accordingly, an increase, preferably, indicates that the multiple sclerosis ther apy is successful. The present invention, further, relates to a method for diagnosing an active status of multiple sclerosis in a subject comprising the steps of: 3@ determining in a sample of the subject the amount of at least one biomarker selected from the biomarkers listed in Table 3; and b) comparing the amount of the said at least one biomarker to a reference amount, whereby an active status of multiple sclerosis is to be diagnosed. 40 In a preferred embodiment of the aforementioned method, said at least one biomarker is select ed from the group of biomarkers listed in Table 3a and wherein an increase in the said at least one biomarker is indicative for an active status of MS. Said increase is preferably an increase of WO 2012/164026 PCT/EP2012/060262 -19 at least 10%, of at least 20%, of at least 30%, of at least 50% (in an increasing order of prefer ence). In another preferred embodiment of the aforementioned method, said at least one biomarker is 5 selected from the group of biomarkers listed in Table 3b and wherein a decrease in the said at least one biomarker is indicative for an active status of MS. Said decrease is preferably an de crease of at least 10%, of at least 20%, of at least 30%, of at least 50% (in an increasing order of preference). 10 The method for diagnosing the active status of MS in a subject can be, preferably, also applied in order to monitor disease progression. In particular, if the presence of an active status is de termined in all samples taken over a certain time window without a sample indicating a remis sion status, the method will also provide an indication for disease progression, e.g., from the relapsing-remitting subtype to the secondary progressing subtype. Thus, the frequency and 15 numbers of active status which can be diagnosed by the method of the present invention can be used to further diagnose aggressiveness and progression of MS. The present invention also relates to a method for predicting whether a subject is at risk of de veloping multiple sclerosis comprising the steps of: 2) determining in a sample of the subject the amount of at least one ceramide, or the total amount of ceramides, and b) comparing the amount of the said at least one ceramide, or the total amount of ceramides, to a reference amount, whereby it is predicted whether a subject is at risk of developing multiple sclerosis. 25 Preferred ceramides have been described elsewhere herein. Preferably, the ceramide is select ed from the ceramides listed in Table 1 and/or 2. Accordingly, the ceramide in the context of the methods of the present invention is preferably 30 selected from the group of ceramides consisting of ceramide (d16:1,C22:0), ceramide (d16:1,C23:0), ceramide (d16:1,C24:0), ceramide (d17:1,C22:0), ceramide (d17:1,C23:0), ceramide (d17:1,C24:0), ceramide (d17:1,C24:1), ceramide (d18:1,C21:0), ceramide (d18:1,C22:0), ceramide (d18:1,C22:1), ceramide (d18:1,C23:0), ceramide (d18:1,C23:1), ceramide (d18:1, C24:0), ceramide (d18:1, C24:1), ceramide (d18:2,C22:0), ceramide (d18:2, 35 C23:0), ceramide (d18:2,C24:0), and ceramide (d18:2,C24:1). In a further preferred embodiment, the ceramide selected from the group of ceramides consist ing of ceramide (d16:1, C22:0), ceramide (d16:1, C23:0), ceramide (d16:1, C24:0), ceramide (d17:1, C22:0), ceramide (d17:1, C23:0), ceramide (d17:1, C24:0), ceramide (d17:1, C24:1), 40 ceramide (d18:1, C21:0), ceramide (d18:1, C22:0), ceramide (d18:1, C22:1), ceramide (d18:1, C23:0), ceramide (d18:1, C23:1), ceramide (d18:2,C22:0), ceramide (d18:2, C23:0), ceramide WO 2012/164026 PCT/EP2012/060262 - 20 (d18:2, C24:0), and ceramide (d18:2,C24:1). In one embodiment, the ceramide is not ceramide (d18:1, C24:0) or not ceramide (d18:1, C24:1). Moreover, the present invention relates to a method for predicting whether a subject is at risk of 5 developing multiple sclerosis comprising the steps of: a) determining in a sample of the subject the amount of at least one biomarker selected from the biomarkers listed in Table 1 and/or 2 and b) comparing the amount of the said at least one biomarker to a reference amount, whereby it is predicted whether a subject is at risk of developing multiple sclerosis. 10 The term "predicting" as used herein, in general, refers to determining the probability according to which a subject will develop a medical condition or its accompanying symptoms within a cer tain time window after the sample has been taken (i.e. the predictive window). It will be under stood that such a prediction will not necessarily be correct for all (100%) of the investigated sub 15 jects. However, it is envisaged that the prediction will be correct for a statistically significant por tion of subjects of a population of subjects (e.g., the subjects of a cohort study). Whether a por tion is statistically significant can be determined by statistical techniques set forth elsewhere herein. 20 In a preferred embodiment of the aforementioned method for predicting whether a subject is at risk of developing multiple sclerosis, the method is repeated with one or more further samples of the subject which have been taken after the above mentioned (first) sample was taken. Accord ingly, by repeating the prediction several times after the initial prediction was made, the predic tion power of the method can be further increased. 25 Moreover, the present invention relates to a method for predicting whether a subject is at risk of developing an active status of multiple sclerosis. Said method shall comprise the steps of: a) determining in a sample of the subject the amount of at least one ceramide, or the total amount of ceramides, and 89 comparing the amount of the said at least one ceramide, or the total amount of ceramides, to a reference amount, whereby it is predicted whether a subject is at risk of developing an active status of multiple sclerosis. The term "ceramide" has been described elsewhere herein. In the context of the aformentioned 35 method, it is particularly preferred that the ceramide is selected from the ceramides listed in Table 3. Preferably an increased amount of the at least one ceramide (or an increased total amount of ceramides) in comparision to the reference amount is indicative for a subject being at risk of developing an active status of multiple sclerosis. 40 A method for predicting whether a subject is at risk of developing an active status of multiple sclerosis is also envisaged by the present invention. Said method shall comprise the steps of: WO 2012/164026 PCT/EP2012/060262 -21 a) determining in a sample of the subject the amount of at least one biomarker selected from the biomarkers listed in Table 3 and b) comparing the amount of the said at least one biomarker to a reference amount, whereby it is predicted whether a subject is at risk of developing an active status of multiple sclerosis. 5 Furthermore, the present invention relates to a method for identifying whether a subject is in need for a therapy against the active status of multiple sclerosis comprising the steps of the aforementioned two methods for predicting whether a subject is at risk of developing an active status of multiple sclerosis and the further steps of identifying a subject in need if the subject is 10 predicted to be at risk of developing an active status of multiple sclerosis. The invention also relates to a method for establishing an aid for diagnosing whether a subject suffering from MS, or not, or the active status of MS, or not, or is at risk of developing MS or the active status thereof is contemplated, said method comprising: a) 5 determining the amount of at least one biomarker as referred to above in a sample of said subject, said determining comprises (i) bringing the sample into contact with a detection agent that specifically binds to said at least one biomarker for a time sufficient to allow for the for mation of a complex of the said detection agent and the biomarker from the sample, (ii) measur ing the amount of the formed complex, wherein the said amount of the formed complex is pro 20 portional to the amount of biomarker present in the sample, and (iii) transforming the amount of the formed complex into an amount of biomarker reflecting the amount of the biomarker present in the sample; b) comparing said amount to a reference; and c) establishing an aid for diagnosing whether a subject suffering from MS, or not, or the active 25 status of MS, or not, or is at risk of developing MS or the active status thereof based on the re sult of the comparison made in step b). A suitable detection agent may be, preferably, an antibody which is specifically binds to the at least one biomarker in a sample of a subject to be investigated by the method of the invention. 30 Another detection agent that can be applied, preferably, may be an aptamere which specifically binds to at least one biomarker in the sample. In yet a preferred embodiment, the sample is re moved from the complex formed between the detection agent and the at least one biomarker prior to the measurement of the amount of formed complex. Accordingly, in a preferred embod iment, the detection agent may be immobilized on a solid support. In yet a preferred embodi 35 ment, the sample can be removed from the formed complex on the solid support by applying a washing solution. The formed complex shall be proportional to the amount of the at least one biomarker present in the sample. It will be understood that the specificity and/or sensitivity of the detection agent to be applied defines the degree of proportion of at least one biomarker com prised in the sample which is capable of being specifically bound. Further details on how the 40 determination can be carried out are also found elsewhere herein. The amount of formed com plex shall be transformed into an amount of at least one biomarker reflecting the amount indeed present in the sample. Such an amount, preferably, may be essentially the amount present in WO 2012/164026 PCT/EP2012/060262 - 22 the sample or may be, preferably, an amount which is a certain proportion thereof due to the relationship between the formed complex and the amount present in the original sample. In yet a preferred embodiment of the aforementioned method, step a) may be carried out by an 5 analyzing unit, in an aspect, an analyzing unit as defined elsewhere herein. In a preferred embodiment of the method of the invention, the amount determined in step a) is compared to a reference. Preferably, the reference is a reference as defined elsewhere herein. In yet another preferred embodiment, the reference takes into account the proportional relation 10 ship between the measured amount of complex and the amount present in the original sample. Thus, the references applied in a preferred embodiment of the method of the invention are arti ficial references which are adopted to reflect the limitations of the detection agent that has been used. In another preferred embodiment, said relationship can be also taken into account when carrying out the comparison, e.g., by including a normalization and/or correction calculation step 15 for the determined amount prior to actually comparing the value of the determined amount and the reference. Again, the normalization and/or correction calculation step for the determined amount adopts the comparison step such that the limitations of the detection agent that has been used are reflected properly. Preferably, the comparison is carried out automatically, e.g., assisted by a computer system or the like. 20 The aid for diagnosing is established based on the comparison carried out in step b) by allocat ing the subject either into a group of subjects suffering MS with certain likelihood or a group of subjects not suffering therefrom. As discussed elsewhere herein already, the allocation of the investigated subject must not be correct in 100% of the investigated cases. Moreover, the 25 groups of subjects into which the investigated subject is allocated are artificial groups in that they are established based on statistical considerations, i.e. a certain preselected degree of likelihood based on which the method of the invention shall operate. Thus, the method may es tablish an aid of diagnosis which may, in an aspect, require further strengthening of the diagno sis by other techniques. Preferably, the aid for diagnosing is established automatically, e.g., 30 assisted by a computer system or the like. In a preferred embodiment of the method of the invention, the determination of the at least one biomarker is achieved by mass spectroscopy techniques (preferably GCMS and/or LCMS), NMR or others referred to herein above. In such cases, preferably, the sample to be analyzed is 35 pretreated. Said pretreatment, preferably, includes obtaining of the at least one biomarker from sample material, e.g., plasma or serum may be obtained from whole blood or the at least one biomarker may even be specifically extracted from sample material. Moreover, for GCMS, fur ther sample pretreatment such as derivatization of the at least one biomarker is, preferably, re quired. Furthermore, pretreatment also, preferably, includes diluting sample material and adjust 40 ing or normalizing the concentration of the components comprised therein. To this end, prefera bly, normalization standards may be added to the sample in predefined amounts which allow for WO 2012/164026 PCT/EP2012/060262 - 23 making a comparison of the amount of the at least one biomarker and the reference and/or be tween different samples to be analyzed. In a preferred embodiment of the method of the invention, said method further comprises a step 5 of recommending and/or managing the subject according to the result of the aid of diagnosis established in step c). Such a recommendation may, in an aspect, be an adaptation of life style, nutrition and the like aiming to improve the life circumstances, the application of therapeutic measures as set forth elsewhere herein in detail, and/or a regular disease monitoring. 10 In another preferred embodiment of the aforementioned method, steps b) and/or c) are carried out by an evaluation unit as set forth elsewhere herein. The method, in another preferred embodiment, also includes a step of managing or treating a subject according to the recommendation or diagnostic result. Preferably, said treating encom 15 passes administering to the subject a therapeutically effective dose of at least one drug selected from the group consisting of: Interferon Betala, Interferon Beta 1b, Azathioprin, Cyclophos phamide, Glatiramer Acetate, Immunglobuline Methotrexat, Mitoxantrone, Leustatin, IVIg, Na talizumab, Teriflunomid, Statins, Daclizumab, Alemtuzumab, Ritximab, Sphingosin 1 phosphate antagonist Fingolimod (FTY720), Cladribine, Fumarate, Laquinimod, drugs affecting B-cells, 20 and antisense agents and agents against CD49d. Accordingly, the present invention also contemplates a method of treating MS in a subject suf fering thereof comprising (a) diagnosing or predicting MS or the active status of MS in a subject by applying a method as referred to herein above and (b) administering to a subject which is 25 identified to suffer from MS or the active status of MS or which is at risk of developing MS or the active status of MS a therapeutically effective amount of at least one drug selected from the group consisting of: Interferon Beta1a, Interferon Beta 1b, Azathioprin, Cyclophosphamide, Glatiramer Acetate, Immunglobuline Methotrexat, Mitoxantrone, Leustatin, IVIg, Natalizumab, Teriflunomid, Statins, Daclizumab, Alemtuzumab, Ritximab, Sphingosin 1 phosphate antagonist 30 Fingolimod (FTY720), Cladribine, Fumarate, Laquinimod, drugs affecting B-cells, and antisense agents and agents against CD49d. The aforementioned methods for the determination of the at least one biomarker can be imple mented into a device. A device as used herein shall comprise at least the aforementioned 35 means. Moreover, the device, preferably, further comprises means for comparison and evalua tion of the detected characteristic feature(s) of the at least one biomarker and, also preferably, the determined signal intensity. The means of the device are, preferably, operatively linked to each other. How to link the means in an operating manner will depend on the type of means included into the device. For example, where means for automatically qualitatively or quantita 40 tively determining the biomarker are applied, the data obtained by said automatically operating means can be processed by, e.g., a computer program in order to facilitate the assessment. Preferably, the means are comprised by a single device in such a case. Said device may ac- WO 2012/164026 PCT/EP2012/060262 - 24 cordingly include an analyzing unit for the biomarker and a computer unit for processing the resulting data for the assessment. Preferred devices are those which can be applied without the particular knowledge of a specialized clinician, e.g., electronic devices which merely require loading with a sample. 5 Alternatively, the methods for the determination of the at least one biomarker can be imple mented into a system comprising several devices which are, preferably, operatively linked to each other. Specifically, the means must be linked in a manner as to allow carrying out the method of the present invention as described in detail above. Therefore, operatively linked, as 10 used herein, preferably, means functionally linked. Depending on the means to be used for the system of the present invention, said means may be functionally linked by connecting each mean with the other by means which allow data transport in between said means, e.g., glass fiber cables, and other cables for high throughput data transport. Nevertheless, wireless data transfer between the means is also envisaged by the present invention, e.g., via LAN (Wireless 15 LAN, W-LAN). A preferred system comprises means for determining biomarkers. Means for determining biomarkers as used herein encompass means for separating biomarkers, such as chromatographic devices, and means for metabolite determination, such as mass spectrometry devices. Suitable devices have been described in detail above. Preferred means for compound separation to be used in the system of the present invention include chromatographic devices, 20 more preferably devices for liquid chromatography, HPLC, and/or gas chromatography. Pre ferred devices for compound determination comprise mass spectrometry devices, more prefer ably, GC-MS, LC-MS, direct infusion mass spectrometry, FT-ICR-MS, CE-MS, HPLC-MS, quad rupole mass spectrometry, sequentially coupled mass spectrometry (including MS-MS or MS MS-MS), ICP-MS, Py-MS or TOF. The separation and determination means are, preferably, 25 coupled to each other. Most preferably, LC-MS and/or GC-MS are used in the system of the present invention as described in detail elsewhere in the specification. Further comprised shall be means for comparing and/or analyzing the results obtained from the means for determination of biomarkers. The means for comparing and/or analyzing the results may comprise at least one databases and an implemented computer program for comparison of the results. Preferred em 30 bodiments of the aforementioned systems and devices are also described in detail below. Therefore, the present invention relates to a diagnostic device comprising: an analysing unit comprising a detector for at least one biomarker as listed in any one of Tables 1, 1a, 1 b, 2, 2a, 2b, 3, 3a, 3b wherein said analyzing unit is adapted for determining the amount 35 of the said biomarker detected by the detector, and, operatively linked thereto; an evaluation unit comprising a computer comprising tangibly embedded a computer program code for carrying out a comparison of the determined amount of the at least one biomarker and a reference amount and a data base comprising said reference amount as for the said bi omarker whereby a multiple sclerosis in a subject, a subject is in need for a therapy of multiple 40 sclerosis or the success of a multiple sclerosis is identified, or whereby is it differentiated be tween i) a subject showing symptoms of multiple sclerosis and suffering from multiple sclerosis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclero- WO 2012/164026 PCT/EP2012/060262 - 25 sis, if the result of the comparison for the at least one metabolite is essentially identical to the kind of regulation and/or fold of regulation indicated for the respective at least one biomarker in any one of Tables 1, 1 a, 1 b, 2, 2a, 2b, 3, 3a or 3b. lr6an preferred embodiment, the present invention relates to a diagnostic device comprising: an analysing unit comprising a detector for at least one ceramide as listed in any one of Tables 1, 1 a, 2, 2a, 3 or 3a, or a detector for ceramides wherein said analyzing unit is adapted for de termining the amount of the said at least one cermide detected by the detector, and, operatively linked thereto; ) 10 an evaluation unit comprising a computer comprising tangibly embedded a computer program code for carrying out a comparison of the determined amount of the at least one biomarker and a reference amount and a data base comprising said reference amount as for the said bi omarker whereby a multiple sclerosis in a subject, a subject is in need for a therapy of multiple sclerosis or the success of a multiple sclerosis is identified, or whereby is it differentiated be 15 tween i) a subject showing symptoms of multiple sclerosis and suffering from multiple sclerosis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclero sis, if the result of the comparison for the at least ceramide is essentially identical to the kind of regulation and/or fold of regulation indicated for the respective at least one ceramide in any one of Tables 1, 1a, 2, 2a, 3 or 3a. 20 In a preferred embodiment, the device comprises a further database comprising the kind of reg ulation and/or fold of regulation values indicated for the respective at least one biomarker in any one of Tables 1, 1 a, 1 b, 2, 2a, 2b, 3, 3a, 3b and a further tangibly embedded computer program code for carrying out a comparison between the determined kind of regulation and/or fold of 25 regulation values and those comprised by the database. Furthermore, the present invention relates to a data collection comprising characteristic values of at least one biomarker being indicative for a medical condition or effect as set forth above 30 (i.e. diagnosis of multiple sclerosis in a subject, identifying whether a subject is in need for a therapy of multiple sclerosis or determining whether a multiple sclerosis therapy is successful). The term "data collection" refers to a collection of data which may be physically and/or logically grouped together. Accordingly, the data collection may be implemented in a single data storage 35 medium or in physically separated data storage media being operatively linked to each other. Preferably, the data collection is implemented by means of a database. Thus, a database as used herein comprises the data collection on a suitable storage medium. Moreover, the data base, preferably, further comprises a database management system. The database manage ment system is, preferably, a network-based, hierarchical or object-oriented database manage 40 ment system. Furthermore, the database may be a federal or integrated database. More prefer ably, the database will be implemented as a distributed (federal) system, e.g. as a Client Server-System. More preferably, the database is structured as to allow a search algorithm to WO 2012/164026 PCT/EP2012/060262 - 26 compare a test data set with the data sets comprised by the data collection. Specifically, by us ing such an algorithm, the database can be searched for similar or identical data sets being in dicative for a medical condition or effect as set forth above (e.g. a query search). Thus, if an identical or similar data set can be identified in the data collection, the test data set will be asso 5 ciated with the said medical condition or effect. Consequently, the information obtained from the data collection can be used, e.g., as a reference for the methods of the present invention de scribed above. More preferably, the data collection comprises characteristic values of all me tabolites comprised by any one of the groups recited above. 10 In light of the foregoing, the present invention encompasses a data storage medium comprising the aforementioned data collection. The term "data storage medium" as used herein encompasses data storage media which are 15 based on single physical entities such as a CD, a CD-ROM, a hard disk, optical storage media, or a diskette. Moreover, the term further includes data storage media consisting of physically separated entities which are operatively linked to each other in a manner as to provide the aforementioned data collection, preferably, in a suitable way for a query search. 20 The present invention also relates to a system comprising: (a) means for comparing characteristic values of the at least one biomarker of a sample operatively linked to (b) a data storage medium as described above. 25 The term "system" as used herein relates to different means which are operatively linked to each other. Said means may be implemented in a single device or may be physically separated devices which are operatively linked to each other. The means for comparing characteristic val ues of biomarkers, preferably, based on an algorithm for comparison as mentioned before. The 30 data storage medium, preferably, comprises the aforementioned data collection or database, wherein each of the stored data sets being indicative for a medical condition or effect referred to above. Thus, the system of the present invention allows identifying whether a test data set is comprised by the data collection stored in the data storage medium. Consequently, the methods of the present invention can be implemented by the system of the present invention. 35 In a preferred embodiment of the system, means for determining characteristic values of bi omarkers of a sample are comprised. The term "means for determining characteristic values of biomarkers" preferably relates to the aforementioned devices for the determination of metabo lites such as mass spectrometry devices, NMR devices or devices for carrying out chemical or 40 biological assays for the biomarkers.
WO 2012/164026 PCT/EP2012/060262 - 27 Moreover, the present invention relates to a diagnostic means comprising means for the deter mination of at least one biomarker selected from any one of the groups referred to above. The term "diagnostic means", preferably, relates to a diagnostic device, system or biological or 5 chemical assay as specified elsewhere in the description in detail. The expression "means for the determination of at least one biomarker" refers to devices or agents which are capable of specifically recognizing the biomarker. Suitable devices may be spectrometric devices such as mass spectrometry, NMR devices or devices for carrying out 10 chemical or biological assays for the biomarkers. Suitable agents may be compounds which specifically detect the biomarkers. Detection as used herein may be a two-step process, i.e. the compound may first bind specifically to the biomarker to be detected and subsequently generate a detectable signal, e.g., fluorescent signals, chemiluminescent signals, radioactive signals and the like. For the generation of the detectable signal further compounds may be required which 15 are all comprised by the term "means for determination of the at least one biomarker". Com pounds which specifically bind to the biomarker are described elsewhere in the specification in detail and include, preferably, enzymes, antibodies, ligands, receptors or other biological mole cules or chemicals which specifically bind to the biomarkers. 20 Further, the present invention relates to a diagnostic composition comprising at least one bi omarker selected from any one of the groups referred to above. The at least one biomarker selected from any of the aforementioned groups will serve as a bi 25 omarker, i.e. an indicator molecule for a medical condition or effect in the subject as set for the elsewhere herein. Thus, the metabolite molecules itself may serve as diagnostic compositions, preferably, upon visualization or detection by the means referred to in herein. Thus, a diagnostic composition which indicates the presence of a biomarker according to the present invention may also comprise the said biomarker physically, e.g., a complex of an antibody and the me 30 tabolite to be detected may serve as the diagnostic composition. Accordingly, the diagnostic composition may further comprise means for detection of the metabolites as specified else where in this description. Alternatively, if detection means such as MS or NMR based tech niques are used, the molecular species which serves as an indicator for the risk condition will be the at least one biomarker comprised by the test sample to be investigated. Thus, the at least 35 one biomarker referred to in accordance with the present invention shall serve itself as a diag nostic composition due to its identification as a biomarker. In general, the present invention contemplates the use of at least one biomarker selected from 40 the biomarkers listed in any one of Tables 1, 2, 1 a, 2a, 1 b, or 2b in a sample of a subject for the diagnosis of multiple sclerosis, the use of at least one biomarker selected from the biomarkers listed in any one of Tables 3, 3a; or 3b; in a sample of a subject for diagnosing an active status WO 2012/164026 PCT/EP2012/060262 - 28 of multiple sclerosis, or the use of at least one biomarker selected from the biomarkers of Table 1 and/or 2 in a sample of a subject for predicting multiple sclerosis as well as the use of at least one biomarker selected from the biomarkers of Table 3 in a sample of a subject for predicting an active status of multiple sclerosis. 5 All references cited herein are herewith incorporated by reference with respect to their disclo sure content in general or with respect to the specific disclosure contents indicated above. 10 The invention will now be illustrated by the following Examples which are not intended to restrict or limit the scope of this invention. Example 1: Study design for the differential diagnosis of Multiple Sclerosis (MS) from other neu rological symptoms or diseases. 15 Specification of the samples: Cases: 41 newly diagnosed therapy naive patients with MS (n=15) or a Clinical Isolated Syn drom (CIS) (n=26). 20 Controls: 42 patients with different neurological symptoms or diseases not being CIS or MS (e.g. headache (n=11), hypaesthesia (n=10), paresthesia (n=6), meningitis patients (n=4) pa tients' with other symptoms (n=1 1). 25 Of the patients with other symptoms one patient was diagnosed with abducens paresis, one patient had leg ache, one patient suffered from dysarthria, one patient suffered from the Els berg-syndrome, one patient showed idiopathic facial nerve paresis, one patient suffered from headache and hypestesia, one patient suffered from neuropathy, one patient suffered from pseudotumor cerebri, and two patients exhibited giddiness. 30 Of the 41 CIS and MS patients, 18 patients had change in signs or symptoms (acute attack), while 23 did not clinically change (stable phase). Additional information from all subjects (e.g. gender, age, BMI, date of sampling, disease sta tus) was used for analysis as described in Example 3 in detail. 35 Serum was prepared from all samples by centrifugation, and samples were stored at -80 'C until measurements had been performed. Example 2: Determination of metabolites 40 Human serum samples were prepared and subjected to LC-MS/MS and GC-MS or SPE-LC MS/MS (hormones) analysis as described in the following: WO 2012/164026 PCT/EP2012/060262 - 29 Proteins were separated by precipitation from blood serum. After addition of water and a mix ture of ethanol and dichloromethane the remaining sample was fractioned into an aqueous, po lar phase and an organic, lipophilic phase. 5 For the transmethanolysis of the lipid extracts a mixture of 140 pl of chloroform, 37 pl of hydro chloric acid (37% by weight HCI in water), 320 pl of methanol and 20 pl of toluene was added to the evaporated extract. The vessel was sealed tightly and heated for 2 hours at 1 00 0 C, with shaking. The solution was subsequently evaporated to dryness. The residue was dried com pletely. 10 The methoximation of the carbonyl groups was carried out by reaction with methoxyamine hy drochloride (20 mg/ml in pyridine, 100 I] for 1.5 hours at 60'C) in a tightly sealed vessel. 20 pl of a solution of odd-numbered, straight-chain fatty acids (solution of each 0.3 mg/mL of fatty acids from 7 to 25 carbon atoms and each 0.6 mg/mL of fatty acids with 27, 29 and 31 carbon 15 atoms in 3/7 (v/v) pyridine/toluene) were added as time standards. Finally, the derivatization with 100 pl of N-methyl-N-(trimethylsilyl)-2,2,2-trifluoroacetamide (MSTFA) was carried out for 30 minutes at 60'C, again in the tightly sealed vessel. The final volume before injection into the GC was 220 pl. 20 For the polar phase the derivatization was performed in the following way: The methoximation of the carbonyl groups was carried out by reaction with methoxyamine hydrochloride (20 mg/ml in pyridine, 50 pl for 1.5 hours at 60'C) in a tightly sealed vessel. 10 pl of a solution of odd numbered, straight-chain fatty acids (solution of each 0.3 mg/mL of fatty acids from 7 to 25 car bon atoms and each 0.6 mg/mL of fatty acids with 27, 29 and 31 carbon atoms in 3/7 (v/v) pyri 25 dine/toluene) were added as time standards. Finally, the derivatization with 50 pl of N-methyl-N (trimethylsilyl)-2,2,2-trifluoroacetamide (MSTFA) was carried out for 30 minutes at 60'C, again in the tightly sealed vessel. The final volume before injection into the GC was 110 pl. The GC-MS systems consist of an Agilent 6890 GC coupled to an Agilent 5973 MSD. The au 30 tosamplers are CompiPal or GCPal from CTC. For the analysis usual commercial capillary separation columns (30 m x 0,25 mm x 0,25 pm) with different poly-methyl-siloxane stationary phases containing 0 % up to 35% of aromatic moi 35 eties, depending on the analysed sample materials and fractions from the phase separation step, were used (for example: DB-1ms, HP-5ms, DB-XLB, DB-35ms, Agilent Technologies). Up to 1 pL of the final volume was injected splitless and the oven temperature program was started at 70 'C and ended at 340 'C with different heating rates depending on the sample material and fraction from the phase separation step in order to achieve a sufficient chromatographic separa 40 tion and number of scans within each analyte peak. Furthermore RTL (Retention Time Locking, Agilent Technologies) was used for the analysis and usual GC-MS standard conditions, for ex ample constant flow with nominal 1 to 1.7 ml/min. and helium as the mobile phase gas, ionisa- WO 2012/164026 PCT/EP2012/060262 - 30 tion was done by electron impact with 70 eV, scanning within a m/z range from 15 to 600 with scan rates from 2.5 to 3 scans/sec and standard tune conditions. The H PLC-MS systems consisted of an Agilent 1100 LC system (Agilent Technologies, Wald 5 bronn, Germany) coupled with an API 4000 Mass spectrometer (Applied Biosystem/MDS SCI EX, Toronto, Canada). HPLC analysis was performed on commercially available reversed phase separation columns with C18 stationary phases (for example: GROM ODS 7 pH, Thermo Betasil C1 8). Up to 10 pL of the final sample volume of evaporated and reconstituted polar and lipophilic phase was injected and separation was performed with gradient elution using metha 10 nol/water/formic acid or acetonitrile/water/formic acid gradients at a flowrate of 200 pL/min. Mass spectrometry was carried out by electrospray ionisation in positive mode for the non-polar fraction and negative mode for the polar fraction using multiple-reaction-monitoring-(MRM) mode and fullscan from 100 - 1000 amu. 15 Steroids and their metabolites were measured by online SPE-LC-MS (Solid phase extraction LC-MS). Catecholamines and their metabolites were measured by online SPE-LC-MS as de scribed by Yamada et al. (J. Anal.Toxicol. (26), 2002, 17-22). For both catecholamines and re lated metabolites and steroids and related metabolites, quantification was achieved by means of 20 stable-isotope-labelled standards, and absolute concentrations were calculated. Analysis of complex lipids in serum samples: Total lipids were extracted from serum by liquid/liquid extraction using chloroform/methanol. 25 The lipid extracts were subsequently fractionated by normal phase liquid chromatography (NPLC) into eleven different lipid groups according to Christie (Journal of Lipid Research (26), 1985, 507-512). The lipid classes Monoacylglycerides (MAG), Triacylglycerides (TAG), Phosphatidylcholines 30 (PC), Phosphatidylserines (PS) were measured by GC. The fractions are analyzed by GC-MS after derivatization with TMSH (Trimethyl sulfonium hy droxide), yielding the fatty acid methyl esters (FAME) corresponding to the acyl moieties of the class-separated lipids. The concentrations of FAME from C14 to C24 are determined in each 35 fraction. The fractions were analyzed by LC-MS/MS using electrospray ionization (ESI) and atmospheric pressure chemical ionization (APCI) with detection of specific multiple reaction monitoring (MRM) transitions for cholesterol esters (CE) sphingoymelins (SM), and ceramides (CER) re 40 spectively.
WO 2012/164026 PCT/EP2012/060262 - 31 Sphingosines and sphingosine-1 -phosphates (SP) were analyzed by LC-MS/MS using elec trospray ionization (ESI) with detection of specific multiple reaction monitoring (MRM) transitions as described by Schmidt H et.al., Prostaglandins & other Lipid Mediators 81(2006), 162-170. 5 Metabolites in Tables 1-3 and/or 1 a-3a and/or 1 b-3b derived from one of these fractions include the respective abbreviation in front of their name separated by an underscore. Eicosanoids and related were measured out of serum by offline- and online-SPE LC-MS/MS (Solid phase extraction-LC-MS/MS) (Masoodi M and Nicolaou A: Rapid Commun Mass Spec 10 trom. 2006 20(20): 3023-3029. Absolute quantification was performed by means of stable iso tope-labelled standards. Example 3: Data analysis and statistical evaluation 15 Serum samples were analyzed in randomized analytical sequence design with pooled samples (so called "pool") generated from aliquots of each sample. Following comprehensive analytical validation steps, the raw peak data for each analyte were normalized to the median of pool per analytical sequence to account for process variability (so called "pool-normalized ratios"). If available, absolute concentrations of metabolites were used for statistical analysis. In all other 20 cases, pool-normalized ratios were used. All data were log10-transformed to achieve normal distribution. For the study described in Example 1, a fixed-effects ANOVA model was designed containing the factors age (numerical), BMI (numerical), gender (categorical), sample preparation time 25 (numerical), and diagnosis (all CASES (with or without acute attack), CASES with acute attack, CASES without acute attack, and controls - reference: controls). CASES includes MS-patients as well as CIS-patients. Pairwise interactions of the factors age, BMI, and gender were consid ered due to high number of metabolites influenced by these factors. Statistical significance was read out from p-values of t-statistics. Direction and strength of regulation were obtained on ratio 30 scale by back transformation of contrast estimates ("ratio = 1OAcontrastestimate") by calcula tion of ratios of median values for the groups to be compared. Regulation type was determined for each metabolite as "up" for increased (ratios >1) within the respective group (e.g. CASES with and without acute attacks) vs. reference (controls) and "down" for decreased (ratios <1) vs. reference. All metabolite Tables were filtered for significance (p-value < 0.05). 35 In order to identify biomarkers for multiple sclerosis, the read-out for diagnosis the comparison of all CASES (with or without acute attacks) vs. control reference was considered (Table 1). In order to read out diagnosis effects related periods of acute attacks during multiple sclerosis dis ease CASES with acute attacks were compared with the reference control (Table 2) and fur 40 thermore, the comparison between CASES with acute attacks vs. CASES without acute attacks were analyzed (Table 3).
WO 2012/164026 PCT/EP2012/060262 - 32 The results of the analyses are summarized in the following Tables, below. The biomarkers to be determined in accordance with the methods of the present invention are listed in the follow ing Tables. Biomarkers not precisely defined by their name are further characterized in Table 4. 5 Table 1: Metabolites which are altered between CASES (CIS and MS patients) and Controls (patients with neurological symptoms or diseases) Metabolite Kind of reg- Median of p ulation ("up" CASES value or "down") relative to of t controls test CER-Ceramide (d16:1,C22:0) up 1.22 0.028 CER-Ceramide (d16:1,C23:0) up 1.28 0.017 CER-Ceramide (d16:1,C24:0) up 1.25 0.018 CER-Ceramide (d17:1,C22:0) up 1.23 0.023 CER-Ceramide (d17:1,C23:0) up 1.26 0.015 CER-Ceramide (d17:1,C24:0) up 1.27 0.012 CER-Ceramide (d18:1,C21:0) up 1.18 0.012 CER-Ceramide (d18:1,C22:0) up 1.19 0.010 CER-Ceramide (d18:1,C23:0) up 1.21 0.007 CER-Ceramide (d18:1,C23:1) up 1.18 0.024 CER-Ceramide (d18:2,C22:0) up 1.20 0.018 CER-Ceramide (d18:2,C23:0) up 1.23 0.009 CER-Ceramide (d18:2,C24:0) up 1.19 0.017 CER-Ceramide (d18:2,C24:1) up 1.16 0.016 CER-Ceramide (d18:1,C24:0) up 1.14 0.038 Threonic acid up 1.13 0.022 Phosphatidylcholine (C18:0,C22:6) down 0.87 0.009 Phosphatidylcholine (C16:0,C22:6) down 0.94 0.046 Phosphatidylcholine (C18:2,C20:4) down 0.94 0.046 MAGPalmitic acid (C16:0) down 0.80 0.012 PCDocosahexaenoic acid (C22:cis[4,7,10,13,16,19]6) down 0.85 0.044 10 15 WO 2012/164026 PCT/EP2012/060262 - 33 Table 1a: Metabolites which are increased in CASES (CIS and MS patients) compared to Con trols (patients with neurological symptoms or diseases) Metabolite Kind of regu- Median of p-value lation - "up" CASES of t-test relative to controls CER-Ceramide (d16:1,C22:0) up 1.22 0.028 CER-Ceramide (d16:1,C23:0) up 1.28 0.017 CER-Ceramide (d16:1,C24:0) up 1.25 0.018 CER-Ceramide (d17:1,C22:0) up 1.23 0.023 CER-Ceramide (d17:1,C23:0) up 1.26 0.015 CER-Ceramide (d17:1,C24:0) up 1.27 0.012 CER-Ceramide (d18:1,C21:0) up 1.18 0.012 CER-Ceramide (d18:1,C22:0) up 1.19 0.010 CER-Ceramide (d18:1,C23:0) up 1.21 0.007 CER-Ceramide (d18:1,C23:1) up 1.18 0.024 CER-Ceramide (d18:2,C22:0) up 1.20 0.018 CER-Ceramide (d18:2,C23:0) up 1.23 0.009 CER-Ceramide (d18:2,C24:0) up 1.19 0.017 CER-Ceramide (d18:2,C24:1) up 1.16 0.016 CER-Ceramide (d18:1,C24:0) up 1.14 0.038 Threonic acid up 1.13 0.022 5 Table 1 b: Metabolites which are decreased in CASES (CIS and MS patients) compared to Con trols (patients with neurological symptoms or diseases) Metabolite Kind of Median of p-value regulation CASES of t-test - "down" relative to controls Phosphatidylcholine (C18:0,C22:6) down 0.87 0.009 Phosphatidylcholine (C16:0,C22:6) down 0.94 0.046 Phosphatidylcholine (C18:2,C20:4) down 0.94 0.046 MAGPalmitic acid (C16:0) down 0.80 0.012 PCDocosahexaenoic acid (C22:cis[4,7,10,13,16,19]6) down 0.85 0.044 10 WO 2012/164026 PCT/EP2012/060262 - 34 Table 2: Metabolites which are altered between CASES (CIS and MS patients) with acute attack and Controls (patients with neurological symptoms or diseases) Metabolite Kind of Median of p-value of regulation CASES t-test ("up" or with "down") acute attack relative to controls CER-Ceramide (d16:1,C22:0) up 1.29 0.030 CER-Ceramide (d16:1,C23:0) up 1.35 0.025 CER-Ceramide (d16:1,C24:1) up 1.27 0.025 CER-Ceramide (d17:1,C23:0) up 1.29 0.038 CER-Ceramide (d17:1,C24:1) up 1.24 0.032 CER-Ceramide (d18:1,C21:0) up 1.29 0.003 CER-Ceramide (d18:1,C22:1) up 1.29 0.004 CER-Ceramide (d18:1,C23:0) up 1.24 0.021 CER-Ceramide (d18:1,C23:1) up 1.31 0.005 CER-Ceramide (d18:2,C22:0) up 1.24 0.033 CER-Ceramide (d18:2,C23:0) up 1.28 0.015 CER-Ceramide (d18:2,C24:1) up 1.22 0.016 CER-Ceramide (d18:1,C24:0) up 1.14 0.12 CER-Ceramide (d18:1,C24:1) up 1.12 0.056 Threonic acid up 1.19 0.016 MAGMyristic acid (C14:0) down 0.26 0.008 PSMyristic acid (C14:0) down 0.17 0.048 TAGdihomo-gamma-Linolenic acid (C20:cis[8,11,14]3) down 0.58 0.046 Homovanillic acid (HVA) down 0.75 0.009 Pyruvate up 1.21 0.040 5 10 WO 2012/164026 PCT/EP2012/060262 - 35 Table 2a: Metabolites which are increased in CASES (CIS and MS patients) with acute attack compared to Controls (patients with neurological symptoms or diseases) Metabolite Kind of Median of p-value of regulation - CASES t-test "up" with acute attack relative to controls CER-Ceramide (d16:1,C22:0) up 1.29 0.030 CER-Ceramide (d16:1,C23:0) up 1.35 0.025 CER-Ceramide (d16:1,C24:1) up 1.27 0.025 CER-Ceramide (d17:1,C23:0) up 1.29 0.038 CER-Ceramide (d17:1,C24:1) up 1.24 0.032 CER-Ceramide (d18:1,C21:0) up 1.29 0.003 CER-Ceramide (d18:1,C22:1) up 1.29 0.004 CER-Ceramide (d18:1,C23:0) up 1.24 0.021 CER-Ceramide (d18:1,C23:1) up 1.31 0.005 CER-Ceramide (d18:2,C22:0) up 1.24 0.033 CER-Ceramide (d18:2,C23:0) up 1.28 0.015 CER-Ceramide (d18:2,C24:1) up 1.22 0.016 CER-Ceramide (d18:1,C24:0) up 1.14 0.12 CER-Ceramide (d18:1,C24:1) up 1.12 0.056 Threonic acid up 1.19 0.016 Pyruvate up 1.21 0.040 Table 2b: Metabolites which are decreased in CASES (CIS and MS patients) with acute attack 5 compared to Controls (patients with neurological symptoms or diseases) Metabolite Kind of Median of p-value of regulation CASES t-test ("up" or with "down") acute attack relative to controls MAGMyristic acid (C14:0) down 0.26 0.008 PSMyristic acid (C14:0) down 0.17 0.048 TAG dihomo-gamma-Linolenic acid (C20:cis[8,11,14]3) down 0.58 0.046 Homovanillic acid (HWA) down 0.75 0.009 WO 2012/164026 PCT/EP2012/060262 - 36 Table 3: Metabolites which are altered between CASES (CIS and MS patients) with acute attack and Cases without acute attack Metabolite Kind of Median p-value regulation of CAS- of t-test ("up" or ES with "down") acute attack relative to CASES without attack MAGMyristic acid (C14:0) down 0.14 0.001 CE-Cholesterylester C12:0 up 1.61 0.022 CE-Cholesterylester C18:1 up 1.17 0.012 CER-Ceramide (d18:1,C14:0) up 1.21 0.047 CER-Ceramide (d18:1,C16:0) up 1.14 0.049 CER-Ceramide (d18:1,C18:0) up 1.19 0.046 CER-Ceramide (d18:1,C22:1) up 1.30 0.008 PCDocosapentaenoic acid (C22:cis[7,10,13,16,19]5) up 1.26 0.037 TAGdihomo-gamma-Linolenic acid (C20:cis[8,11,14]3) down 0.36 0.001 TAGDocosapentaenoic acid (C22:cis[4,7,10,13,16]5) down 0.31 0.007 TAG_Eicosenoic acid (C20:cis[ 11]1) down 0.61 0.038 beta-Carotene up 1.33 0.050 Homovanillic acid (HWA) down 0.74 0.014 Isocitrate up 1.15 0.036 Phosphatidylcholine No 02 (1*) up 1.10 0.019 Sphingosine-1-phosphate (d16:1) up 1.23 0.039 TAG (C16:0,C18:2) (*1) down 0.70 0.007 TAG (C18:2,C18:2) (*1) down 0.65 0.004 TAG (C16:0,C18:2,C18:2) down 0.68 0.009 TAG (C16:1,C18:1,C18:2) down 0.68 0.009 5 (1*: see Table4) 10 WO 2012/164026 PCT/EP2012/060262 - 37 Table 3a: Metabolites which are increased in CASES (CIS and MS patients) with acute attack compared to CASES (CIS and MS patients) without acute attack Metabolite Kind of Median of p-value of regulation - CASES t-test "up" with acute attack relative to CASES without attack CE-Cholesterylester C12:0 up 1.61 0.022 CE-Cholesterylester C18:1 up 1.17 0.012 CER-Ceramide (d18:1,C14:0) up 1.21 0.047 CER-Ceramide (d18:1,C16:0) up 1.14 0.049 CER-Ceramide (d18:1,C18:0) up 1.19 0.046 CER-Ceramide (d18:1,C22:1) up 1.30 0.008 PCDocosapentaenoic acid (C22:cis[7,10,13,16,19]5) up 1.26 0.037 beta-Carotene up 1.33 0.050 Isocitrate up 1.15 0.036 Phosphatidylcholine No 02 (1*) up 1.10 0.019 Sphingosine-1-phosphate (d16:1) up 1.23 0.039 (1*: see Table4) 5 Table 3b: Metabolites which are decreased in CASES (CIS and MS patients) with acute attack compared to CASES (CIS and MS patients) without acute attack Metabolite Kind of Median of p-value of regulation - CASES t-test "down" with acute attack relative to CASES without attack MAGMyristic acid (C14:0) down 0.14 0.001 TAGdihomo-gamma-Linolenic acid (C20:cis[8,11,14]3) down 0.36 0.001 TAGDocosapentaenoic acid (C22:cis[4,7,10,13,16]5) down 0.31 0.007 TAG_Eicosenoic acid (C20:cis[ 11]1) down 0.61 0.038 Homovanillic acid (HWA) down 0.74 0.014 TAG (C16:0,C18:2) (*1) down 0.70 0.007 WO 2012/164026 PCT/EP2012/060262 - 38 TAG (C18:2,C18:2) (*1) down 0.65 0.004 TAG (C16:0,C18:2,C18:2) down 0.68 0.009 TAG (C16:1,C18:1,C18:2) down 0.68 0.009 Table 4: Additional chemical/physical properties of biomarkers marked with (*1) in the Tables above Metabolite name Description Phosphatidylcholine No 02 represents the sum parameter of phosphatidylcholines. It exhibits the following characteristic ionic Phosphatidylcholine No 02 species when detected with LC/MS, applying electro-spray ioni zation (ESI) mass spectrometry: mass-to-charge ratio (m/z) of the positively charged ionic species is 808.4 (+/- 0.5). TAG (C16:0,C18:2) represents the sum parameter of triacyl glycerides containing the combination of a C16:0 fatty acid unit and a C18:2 fatty acid unit. It exhibits the following characteristic ionic species when detected with LC/MS, applying electro-spray ionization (ESI) mass spectrometry: mass-to-charge ratio (m/z) of the positively charged ionic species is 575.4 (+/- 0.5). TAG (C18:2,C18:2) represents the sum parameter of triacyl glycerides containing the combination of two C18:2 fatty acid TAG (C18:2,C18:2) units. It exhibits the following characteristic ionic species when detected with LC/MS, applying electro-spray ionization (ESI) mass spectrometry: mass-to-charge ratio (m/z) of the positively charged ionic species is 599.6 (+/- 0.5). 5

Claims (20)

1. A method for diagnosing multiple sclerosis (MS) in a subject comprising the steps of: a) determining in a sample of the subject an amount of at least one biomarker 5 selected from the biomarkers listed in Table 1 and/or Table 2; and b) comparing the amount of the said at least one biomarker to a reference amount, wherein multiple sclerosis is to be diagnosed.
2. The method of claim 1, wherein the said at least one biomarker is selected from the 0 group of biomarkers listed in Table 1a and/or Table 2a and wherein an increase in the said at least one biomarker as compared to the reference amount is indicative for multiple sclerosis.
3. The method of claim 1, wherein the said at least one biomarker is selected from the 5 group of biomarkers listed in Table 1 b and/or Table 2b and wherein a decrease in the said at least one biomarker as compared to the reference amount is indicative for multiple sclerosis.
4. The method of any one of claims 1 to 3, wherein multiple sclerosis is diagnosed in an .0 early stage of multiple sclerosis.
5. The method of any one of claims I to 4, wherein said reference amount is derived from an apparently healthy subject or from a group thereof, or from a subject known not to suffer from multiple sclerosis, but showing multiple sclerosis symptoms, or from .5 a group thereof.
6. A method for identifying whether a subject is in need of a therapy for multiple sclerosis comprising the method of any one of claims I to 5 and further comprising the step of identifying a subject in need of a therapy for multiple sclerosis. 30
7. A method for differentiating between i) a subject showing symptoms of multiple sclerosis and suffering from multiple sclerosis and ii) a subject showing multiple sclerosis symptoms, but not suffering from multiple sclerosis, comprising a) determining in a sample of a subject showing symptoms of multiple 35 sclerosis an amount of at least one biomarker selected from the biomarkers listed in Table 1 and/or Table 2, and b) comparing the amount of the said at least one biomarker to at least one reference amount, wherein it is differentiated between i) and ii). 40
8. A method for determining whether a multiple sclerosis therapy is successful comprising the steps of: 4959563_1 (GHMatters) P95279.AU 16-Dec-13 -40 a) determining at least one biomarker selected from the biomarkers listed in Table I and/or Table 2 in a first and a second sample of the subject wherein said first sample has been taken prior to or at the onset of the multiple sclerosis therapy and said second sample has been taken after 5 the onset of the said therapy; and b) comparing the amount of the said at least one biomarker in the first sample to the amount in the second sample, wherein a change in the amount determined in the second sample in comparison to the first sample is indicative for multiple sclerosis therapy being successful. 0
9. The method of claim 8, wherein: i) said change is a decrease and wherein said at least one biomarker is selected from the biomarkers listed in Table 1 a and/or Table 2a; or ii) said change is an increase and wherein said at least one biomarker is 5 selected from the biomarkers listed in Table 1b and/or Table 2b.
10. The method of claim 8 or claim 9, wherein said therapy comprises administering at least one drug selected from the group consisting of Interferon Beta 1a, Interferon Beta 1 b, Azathioprin, Cyclophosphamide, Glatiramer Acetate, Immunglobuline Methotrexat, 0 Mitoxantrone, Leustatin, IVIg, Natalizumab, Teriflunomid, Statins, Daclizumab, Alemtuzumab, Rituximab, Sphingosin I phosphate antagonist Fingolimod (FTY720), Cladribine, Fumarate, Laquinimod, a drug affecting B-cells, an antisense agent, and an agent against CD49d. !5
11. A method for diagnosing an active status of multiple sclerosis in a subject comprising the steps of: a) determining in a sample of the subject the amount of at least one biomarker selected from the biomarkers listed in Table 3; and b) comparing the amount of the said at least one biomarker to a reference 30 amount, wherein multiple sclerosis is to be diagnosed.
12. The method of claim 11, wherein i) the said at least one biomarker is selected from the group of biomarkers listed in Table 3a and wherein an increase in the said at least one 35 biomarker is indicative for an active status of multiple sclerosis, or ii) the said at least one biomarker is selected from the group of biomarkers listed in Table 3b and wherein a decrease in the said at least one biomarker is indicative for an active status of multiple sclerosis. 40
13. The method of claim 11 or claim 12, wherein said reference amount is derived from a subject or a group of subjects exhibiting a stable status of multiple sclerosis. 49595631 (GHMatters) P95279.AU 16.-Dec-13 -41
14. A method for predicting whether a subject is at risk of developing multiple sclerosis comprising the steps of: a) determining in a sample of the subject an amount of at least one biomarker 5 selected from the biomarkers listed in Table 1 and/or Table 2; and b) comparing the amount of the said at least one biomarker to a reference amount, wherein it is predicted whether a subject is at risk of developing multiple sclerosis. 0
15. A method for predicting whether a subject is at risk of developing an active status of multiple sclerosis comprising the steps of: a) determining in a sample of the subject an amount of at least one biomarker selected from the biomarkers listed in Table 3; and b) comparing the amount of the said at least one biomarker to a reference 5 amount, wherein it is predicted whether a subject is at risk of developing an active status of multiple sclerosis.
16. A diagnostic device comprising: a) an analysing unit comprising a detector for at least one biomarker listed in any one of Tables 1, 1a, 1 b, 2, 2a, 2b, 3, 3a, 3b wherein said analysing unit determines an amount of the said biomarker detected by the detector, and, operatively linked thereto; b) an evaluation unit comprising a computer comprising a tangibly embedded computer program code that compares the determined amount of the at 5 least one biomarker and a reference amount and a database comprising said reference amount as for the said biomarker wherein a multiple sclerosis in a subject, a subject is in need for a therapy of multiple sclerosis or the success of a multiple sclerosis is identified, or wherein it is differentiated between i) a subject showing symptoms of multiple sclerosis 30 and suffering from multiple sclerosis and ii) a subject showing symptoms of multiple sclerosis, but not suffering from multiple sclerosis, if the result of the comparison for the at least one metabolite is essentially identical to the kind of regulation and/or fold of regulation indicated for the respective at least one biomarker in any one of Tables 1, 1a, 1 b, 2, 2a, 2b, 3, 3a or 3b. 35
17. A data collection comprising characteristic values of at least one biomarker listed in any one of Tables 1, 1a, 1 b, 2, 2a, 2b, 3, 3a, 3b being indicative for: diagnosis of multiple sclerosis whether a subject is in need of a therapy for multiple sclerosis; or whether a multiple sclerosis therapy is successful. 40
18. A data storage medium comprising the data collection of claim 17.
4959563.1 (GHMatters) P95279.AU 16-Dec-13 - 42
19. A system comprising: (a) means that compares characteristic values of at least one biomarker of a sample operatively linked to 5 (b) the data storage medium of claim 18.
20. A diagnostic means comprising means that determines at least one biomarker selected from any one of Tables 1, 1 a, 1 b, 2, 2a, 2b, 3, 3a or 3b. 4959503_1 (GHMatter) P95279.AU 16-Dec-13
AU2012264688A 2011-05-31 2012-05-31 Methods for diagnosing multiple sclerosis Abandoned AU2012264688A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161491353P 2011-05-31 2011-05-31
EP11168266.2 2011-05-31
US61/491,353 2011-05-31
EP11168266 2011-05-31
PCT/EP2012/060262 WO2012164026A1 (en) 2011-05-31 2012-05-31 Methods for diagnosing multiple sclerosis

Publications (3)

Publication Number Publication Date
AU2012264688A1 AU2012264688A1 (en) 2013-11-21
AU2012264688A8 AU2012264688A8 (en) 2013-12-19
AU2012264688A2 true AU2012264688A2 (en) 2014-01-23

Family

ID=47258397

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012264688A Abandoned AU2012264688A1 (en) 2011-05-31 2012-05-31 Methods for diagnosing multiple sclerosis

Country Status (5)

Country Link
EP (1) EP2715361A1 (en)
JP (1) JP2014517300A (en)
AU (1) AU2012264688A1 (en)
CA (1) CA2834455A1 (en)
WO (1) WO2012164026A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010326737A1 (en) * 2009-12-01 2012-06-07 Metanomics Health Gmbh Means and methods for diagnosing multiple sclerosis
EP2580596B1 (en) 2010-06-10 2015-02-25 Metanomics Health GmbH Methods to diagnose liver diseases
MA41139A (en) 2014-12-11 2017-10-17 Actelion Pharmaceuticals Ltd PHARMACEUTICAL COMBINATION INCLUDING A SIP1 RECEPTOR SELECTIVE AGONIST
US11061034B2 (en) 2015-10-14 2021-07-13 Public University Corporation Yokohama City University Blood biomarker for use in evaluation of effect of drug therapy on kidney cancer

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4540884A (en) 1982-12-29 1985-09-10 Finnigan Corporation Method of mass analyzing a sample by use of a quadrupole ion trap
US5397894A (en) 1993-05-28 1995-03-14 Varian Associates, Inc. Method of high mass resolution scanning of an ion trap mass spectrometer
WO2003073464A1 (en) 2002-02-28 2003-09-04 Metanomics Gmbh & Co. Kgaa Mass spectrometry method for analysing mixtures of substances
NL1021892C2 (en) * 2002-11-11 2004-05-12 Tno Detection of demyelination.
US8026099B2 (en) * 2007-07-26 2011-09-27 Washington University Lipid profile as a biomarker for early detection of neurological disorders

Also Published As

Publication number Publication date
WO2012164026A8 (en) 2013-04-18
JP2014517300A (en) 2014-07-17
CA2834455A1 (en) 2012-12-06
WO2012164026A1 (en) 2012-12-06
AU2012264688A8 (en) 2013-12-19
EP2715361A1 (en) 2014-04-09
AU2012264688A1 (en) 2013-11-21

Similar Documents

Publication Publication Date Title
AU2017201020B2 (en) Means and methods for diagnosing pancreatic cancer in a subject
US10393762B2 (en) Means and methods for diagnosing heart failure in a subject
US20120238028A1 (en) Means and Methods for Diagnosing Multiple Sclerosis
EP2580596B1 (en) Methods to diagnose liver diseases
JP6219300B2 (en) Apparatus and method for diagnosing pancreatic cancer
EP2369346A2 (en) Means and method for diagnosing diabetes
WO2015028671A1 (en) Means and methods for diagnosing heart failure in a subject
AU2012264688A2 (en) Methods for diagnosing multiple sclerosis

Legal Events

Date Code Title Description
TH Corrigenda

Free format text: IN VOL 27 , NO 45 , PAGE(S) 6475 UNDER THE HEADING PCT APPLICATIONS THAT HAVE ENTERED THE NATIONAL PHASE - NAME INDEX UNDER THE NAME METANOMICS HEALTH GMBH, APPLICATION NO. 2012264688, UNDER INID (71) CORRECT THE APPLICANT NAME TO METANOMICS HEALTH GMBH

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 16 DEC 2013

MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application