WO2008125651A2 - Biomarkers for multiple sclerosis - Google Patents

Biomarkers for multiple sclerosis Download PDF

Info

Publication number
WO2008125651A2
WO2008125651A2 PCT/EP2008/054479 EP2008054479W WO2008125651A2 WO 2008125651 A2 WO2008125651 A2 WO 2008125651A2 EP 2008054479 W EP2008054479 W EP 2008054479W WO 2008125651 A2 WO2008125651 A2 WO 2008125651A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
multiple sclerosis
seq
polypeptide
patients
Prior art date
Application number
PCT/EP2008/054479
Other languages
French (fr)
Other versions
WO2008125651A3 (en
Inventor
Veerle Somers
Pieter Stinissen
Original Assignee
Apitope International Nv
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
Priority to JP2010502532A priority Critical patent/JP2010523629A/en
Priority to CA002683865A priority patent/CA2683865A1/en
Priority to MX2009011007A priority patent/MX2009011007A/en
Priority to EP08749552A priority patent/EP2145182A2/en
Priority to US12/595,597 priority patent/US8772225B2/en
Priority to AU2008237878A priority patent/AU2008237878A1/en
Application filed by Apitope International Nv filed Critical Apitope International Nv
Priority to CN200880018874A priority patent/CN101680883A/en
Publication of WO2008125651A2 publication Critical patent/WO2008125651A2/en
Publication of WO2008125651A3 publication Critical patent/WO2008125651A3/en
Priority to ZA2009/07027A priority patent/ZA200907027B/en
Priority to NO20093327A priority patent/NO20093327L/en
Priority to US14/290,782 priority patent/US20140273033A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis

Definitions

  • the present invention relates to the diagnosis of neurological disorders, more specifically to the diagnosis of multiple sclerosis.
  • a biomarker panel is provided which can be used to detect if a subject has multiple sclerosis. Also described are methods of identification of such biomarkers.
  • MS Multiple sclerosis
  • RR relapsing-remitting
  • PP primary
  • SP secondary
  • MS is considered to be of autoimmune origin and is characterized neuropathologically by variable extents of focal inflammation, demyelination, axonal damage, gliotic scarring and atrophy, but also by remyelination and regeneration in the CNS. This has led, together with the clinical variability, to the concept of MS as a heterogenous disease with respect to four pathogenetic mechanisms of demyelination 1 ' 2 .
  • One of these pathogenetic subtypes is characterized neuropathologically by antibody-dependent immune mechanisms involved in the formation of MS lesions 1 ' 3 .
  • an important role of autoreactive B cells and autoantibodies has been demonstrated 4 .
  • CNS central nervous system
  • MBP myelin basic protein
  • OIND other-inflammatory neurological diseases
  • NIND non-inflammatory neurological diseases
  • MS may diagnose MS in some patients soon after the onset of the illness. In others, however, doctors may not be able to readily identify the cause of the symptoms, leading to years of uncertainty and multiple diagnoses. The vast majority of patients are mildly affected, but in the worst cases, MS can render a person unable to write, speak or walk. Unfortunately, no single laboratory test is yet available to prove or rule out MS. Therefore, there is a great need in the art for improved diagnostic tests for MS.
  • the development of a panel of biomarkers, specific for different pathophysiological mechanisms, will be crucial for the further understanding of the pathogenesis of MS, as well as diagnosis, classification, disease activity, and theranostic applications.
  • Figure 1 Expression profile of novel antigenic targets in normal tissues. Expression patterns are shown for UH-CSF1.4 and UH-CSFP1.7. The lower panel shows a control hybridization with an actin probe. Lane 1 : brain; lane 2: heart; lane 3: skeletal muscle; lane 4: colon; lane 5: thymus; lane 6: spleen; lane 7: kidney; lane 8: liver; lane 9: small intestine; lane 10: placenta; lane 1 1 : lung; lane 12: peripheral blood lymphocytes.
  • FIG. 2 UH-CSFP1.7 and UH-CSFP1.8 protein expression in E.coli.
  • UH-CSFP1.7 was cloned as antigen (6,1 kDa) and the UH-CSFP1.8 protein was cloned as partial * (13,3 kDa) and full-length protein (20,3 kDa) with a 16,7 kDa thioredoxin fusion (His tag) resulting in 22,8 kDa protein for UH-CSFP1.7 and 30 kDa partial and 37 kDa full length band for UH-CSFP1.8
  • FIG. 3 SAS procedure, a. A phage-displayed MS cDNA repertoire is preincubated with MS patient CSF antibodies, b. MS-specific antigens displayed on phage (black) bind to MS-antigen specific patient IgG (black), c. Phage antigen-lgG complexes (black) are captured on a surface coated with polyclonal anti-human IgG (checked), d. Nonrelevant phages are washed away, and CSF-IgG specific phages are eluted. e. Selected phages are used for reinfection of bacteria, f. Selected phages are amplified and used for further rounds of selection.
  • FIG. 4 Solution phase assays demonstrate high affinity and specificity of CSF antibodies to UH-CSFP1.1.
  • the UH-CSFP1.1 peptide was pre-incubated at different dilutions with MS-CSF8 and MS-CSF26, respectively, and subsequently, the remaining immunoreactivity measured by
  • Figure 5 Histogram showing reactivity against UH-CSFP1.1 peptide of 10 random clones tested by ELISA assay. A positive signal was obtained for clone 7.
  • Figure 6 Protein expression of UH-CSFP1.3 and UH-CSFP1.6 (partial).
  • Figure 7 Reactivity of 8 individual CSF samples against UH-CSFP1.1 , UH-CSFP1.2, UH- CSFP1.4, UH-CSFP1.5 and negative control.
  • Figure 8 Antibody reactivity towards UH-CSFP1.6 in serum from 16 randomly selected MS patients, 15 NIND/OIND patients and 16 healthy controls.
  • the horizontal line represents the cut-off value.
  • Biomarkers which can be used for the detection of Multiple Sclerosis (MS) in patients.
  • Biomarkers were isolated with the technology of Serological Antigen Selection (SAS) wherein antigens (i.e. biomarkers) were identified that bind to antibodies present in cerebrospinal fluid (CSF) in patients suffering from Multiple Sclerosis.
  • SAS Serological Antigen Selection
  • CSF cerebrospinal fluid
  • a cDNA phage display library comprising cDNA products derived from MS brain plaques - expressed as a fusion to minor coat protein pVI of filamentous phage M13 - was panned to identify cDNA clones that bind auto-antibodies in CSF specimens from MS patients.
  • the invention provides a composition comprising at least two different polypeptides comprising a sequence represented by any of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
  • a composition is herein also designated as a biomarker or as a biomarker panel.
  • the SEQ ID NO: 1-8 correspond with the translated amino acid sequences of the antigens retrieved by the selection of phage displayed MS cDNA expression library on MS patient cerebrospinal fluid (CSF).
  • CSF cerebrospinal fluid
  • the translation of the insert of UH-CSFP1.1 corresponds with SEQ ID NO: 1 , ...
  • the translation of the insert of UH-CSFP1.8 corresponds with SEQ ID NO: 8 (see table 3).
  • the nucleotide sequences which encode SEQ ID NO: 1-8 are depicted in SEQ ID NO: 9-16 (wherein SEQ ID NO: 9 encodes SEQ ID NO: 1 , ..., and SEQ ID NO: 16 encodes SEQ ID NO: 8).
  • composition comprises at least two different polypeptides wherein such a polypeptide comprises a sequence as depicted by SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7 or 8.
  • a polypeptide present in the composition can also be a protein.
  • SEQ ID NO:8 was cloned as a partial 13.3 kDa protein (protein product as detected using SAS). Since SEQ ID NO: 8 (corresponding with UH-CSFP1.8) is a fragment of the SPAG16 protein (which full length is 37 kDa) the composition can also comprise the full length SPAG16 protein.
  • composition of the invention can also comprise at least two different polypeptides wherein said polypeptides are fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7 or 8. It is envisaged that 5 consecutive amino acids derived from SEQ ID NO: 1-8 are sufficient to be recognized as antigens by the autoantibodies present in for example serum or CSF.
  • composition comprises 8 different polypeptides comprising a sequence selected from SEQ ID NO: 1-8 or 8 different fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
  • the composition comprises 4 different polypeptides comprising a sequence represented by SEQ ID NO: 4, 5, 6 and 7 or 4 different fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 4, 5, 6 and 7.
  • the invention provides the use of a composition of the invention for detecting the presence of specific antibodies to at least one polypeptide present in said composition wherein said antibodies are present in a body fluid of a mammal.
  • the invention provides the use of a composition of the invention for detecting the presence of specific auto-antibodies to at least one polypeptide present in said composition wherein said auto-antibodies are present in a body fluid of a mammal.
  • said use of a composition is an "in vitro" use of a composition.
  • body fluid includes blood, blood serum, blood plasma, saliva, urine, tears, bone marrow fluid, cerebrospinal fluid (CSF), synovial fluid, lymphatic fluid, amniotic fluid, nipple aspiration fluid and the like.
  • Preferred body fluids for analysis are those that are conveniently obtained from patients, particularly preferred body fluids include blood serum, blood plasma and CSF.
  • the invention provides a method for detecting multiple sclerosis in a mammal comprising i) detecting the presence of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the presence or quantity of said antibody indicates that said mammal suffers from multiple sclerosis.
  • the method for detecting multiple sclerosis in a mammal of the present invention is combined with the detection of the MS markers described in US20040043431 and more specifically to the markers described in the claims 5, 6, 7, 8, 9 and 10 of said application.
  • the invention provides a method for evaluating the prognosis/disease severity of multiple sclerosis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the increased or decreased concentration of said antibody indicates the prognosis of multiple sclerosis in said mammal.
  • the invention provides a method for selecting mammals for a specific therapeutic treatment of multiple sclerosis or evaluating the therapeutic treatment of multiple sclerosis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the increased or decreased concentration of said antibody leads to an election of a specific therapeutic treatment of multiple sclerosis in said mammal.
  • said body fluid is CSF.
  • said body fluid is serum.
  • said mammal is a human.
  • the invention provides an antibody that specifically binds to a polypeptide selected from the group comprising of polypeptides selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
  • Methods for generating antibodies are well known in the art.
  • the antibodies are monoclonal antibodies.
  • the polypeptides forming part of the compositions of the invention may be synthesized chemically or may be made in a recombinant way. They may also be coupled to a soluble carrier after synthesis or after recombinant production.
  • a carrier the nature of such a carrier should be such that it has a molecular weight greater than 5000 and should not be recognized by antibodies.
  • a carrier can be a protein. Proteins which are frequently used as carriers are keyhole limpet hemocyanin, bovine gamma globulin, bovine serum albumin, and poly-L-lysine. There are many well described techniques for coupling peptides to carriers. The linkage may occur at the N-terminus, C-terminus or at an internal site in the peptide. The polypeptide may also be derivatized for coupling.
  • polypeptides may also be synthesized directly on an oligo-lysine core in which both the alpha as well as the epsilon-amino groups of lysines are used as growth points for the polypeptides.
  • the number of lysines comprising the core is preferably 3 or 7.
  • a cysteine may be included near or at the C-terminus of the complex to facilitate the formation of homo- or heterodimers.
  • the invention relates to a process for detecting antibodies related to MS in a biological sample of a mammal liable to contain them, this process comprising contacting the biological sample with a composition according to the invention under conditions enabling an immunological reaction between said composition and the antibodies which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed.
  • the detection can be carried out according to any classical process.
  • immunoenzymatic processes according to the ELISA technique or immunofluorescent or radioimmunological (RIA) or the equivalent ones (e.g. LINE blot or LINE assay) can be used.
  • the invention also relates to polypeptides according to the invention labeled by an appropriate label of the enzymatic, fluorescent, biotin, radioactive type.
  • a method for detecting antibodies related to MS comprises for instance the following steps: deposit of determined amounts of a polypeptidic composition according to the invention on a support (e.g. into wells of a titration microplate), introduction on said support (e.g. into wells) of increasing dilutions of the body fluid (e.g. CSF) to be diagnosed, incubation of the support (e.g. microplate), repeated rinsing of the support (e.g.
  • microplate introduction on the support labeled antibodies which are specific for immunoglobulins present in the body fluid, the labeling of these antibodies being based on the activity of an enzyme which is selected from among the ones which are able to hydrolyze a substrate by modifying the absorption of the radiation of this latter at least at a given wave length, detection by comparing with a control standard of the amount of hydrolyzed substrate.
  • the invention also relates to a process for detecting and identifying antigens of MS in a body fluid liable to contain them, this process comprising: contacting the biological sample with an appropriate antibody of the invention (i.e. antibodies with a specificity for a polypeptide of the composition) under conditions enabling an immunological reaction between said antibody and the antigens of MS which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed.
  • an appropriate antibody of the invention i.e. antibodies with a specificity for a polypeptide of the composition
  • antibodies in particular auto-antibodies, which recognize the polypeptides of the invention, can be detected in a variety of ways.
  • One method of detection is further described in the examples and uses enzyme-linked immunosorbant assay (ELISA) of the polypeptides of the invention displayed by phages (i.e. phage-ELISA technology).
  • ELISA enzyme-linked immunosorbant assay
  • phage-ELISA technology i.e. phage-ELISA technology
  • the latter technology is fully described in Somers V. et al (2005) J. of Autoimmunity 25: 223-228, wherein paragraph 2.6 on page 225 is herein specifically incorporated).
  • a polypeptide or a mixture of polypeptides is bound to a solid support.
  • this will be a microtiter plate but may in principle be any sort of insoluble solid phase (e.g. glass, nitrocellulose).
  • a suitable dilution or dilutions of for example CSF or serum to be tested is brought into contact with the solid phase to which the polypeptide is bound.
  • a solution hybridization is carried out in which high affinity interactions occur (eg. biotinylated polypeptides of the composition are pre-incubated with CSF). The incubation is carried out for a time necessary to allow the binding reaction to occur.
  • immune complexes i.e. auto-antibodies present in for example human CSF binding to at least one polypeptide of the invention
  • detection of immune complexes is achieved using antibodies which specifically bind to human immunoglobulins, and which have been labeled with an enzyme, preferably but not limited to either horseradish peroxidase, alkaline phosphatase, or beta-galactosidase, which is capable of converting a colorless or nearly colorless substrate or co-substrate into a highly colored product or a product capable of forming a colored complex with a chromogen.
  • an enzyme preferably but not limited to either horseradish peroxidase, alkaline phosphatase, or beta-galactosidase, which is capable of converting a colorless or nearly colorless substrate or co-substrate into a highly colored product or a product capable of forming a colored complex with a chromogen.
  • the detection system may employ an enzyme which, in the presence of
  • the amount of product formed is detected either visually, spectrophotometrically, electrochemically, fluorescently or luminometrically, and is compared to a similarly treated control.
  • the detection system may also employ radioactively labeled antibodies, in which case the amount of immune complex is quantified by scintillation counting or gamma counting.
  • Other detection systems which may be used include those based on the use of protein A derived from Staphylococcus aureus Cowan strain I, protein G from group C Staphylococcus sp. (strain 26RP66), or systems which make use of the high affinity biotin- avidin or streptavidin binding reaction.
  • polypeptides of the invention may be either labeled or unlabeled. Labels which may be employed may be of any type, such as enzymatic, chemical, fluorescent, luminescent, or radioactive.
  • the polypeptides may be modified for binding to surfaces or solid phases, such as, for example, microtiter plates, nylon membranes, glass or plastic beads, and chromatographic supports such as cellulose, silica, or agarose. The methods by which polypeptides can be attached or bound to solid support or surface are well known to those skilled in the art.
  • polypeptides of the invention can be prepared according to the classical techniques in the field of peptide synthesis.
  • the synthesis can be carried out in homogeneous solution or in solid phase.
  • the synthesis technique in homogeneous solution which can be used is the one described by Houbenweyl in the book titled "Methode der organischen chemie"
  • polypeptides of the invention can also be prepared in solid phase according to the method described by Atherton & Shepard in their book titled “Solid phase peptide synthesis” (Ed. IRL
  • antibodies raised to polypeptides of the invention can also be used in conjunction with labeled polypeptides of the invention for the detection of (auto)-antibodies present in serum or CSF by competition assay.
  • antibodies raised to polypeptides are attached to a solid support which may be, for example, a plastic bead or a plastic tube. Labeled polypeptide is then mixed with suitable dilutions of the fluid (e.g. CSF) to be tested and this mixture is subsequently brought into contact with the antibody bound to the solid support. After a suitable incubation period, the solid support is washed and the amount of labeled polypeptide is quantified.
  • a reduction in the amount of label bound to the solid support is indicative of the presence of (auto)-antibodies in the original sample.
  • the polypeptide may also be bound to the solid support. Labeled antibody may then be allowed to compete with (auto)-antibody present in the sample (e.g. CSF) under conditions in which the amount of polypeptide is limiting. As in the previous example, a reduction in the measured signal is indicative of the presence of (auto)-antibodies in the sample tested.
  • a test for giving evidence of the fact that one or more polypeptides present in a composition of the invention are recognized by antibodies present in for example CSF of serum is an immunoblotting (or Western blotting) analysis.
  • polypeptides can be chemically synthesized or polypeptides (or the protein) can be produced via recombinant techniques.
  • polypeptides of the invention are blotted onto nitrocellulose membranes (e.g. Hybond C. (Amersham)) as described by Towbin H.
  • nitrocellulose sheets are incubated overnight with each of these samples (e.g. diluted 1 :50) (after blocking a-specific protein-binding sites). Reactive areas on the nitrocellulose sheets are revealed by incubation with e.g. peroxidase conjugated goat anti-human immunoglobulin G antibody (e.g. diluted 1 :200) for 4 h, and after repeated washings, color reaction is developed by adding for example alpha-chloronaphtol (Bio-Rad Laboratories, Richmond, Calif.) in the presence of hydrogen peroxide.
  • peroxidase conjugated goat anti-human immunoglobulin G antibody e.g. diluted 1 :200
  • the free reactive functions which are present in some of the amino acids, which are part of the constitution of the polypeptides of the invention particularly the free carboxyl groups which are carried by the groups GIu and Asp or by the C-terminal amino acid on the one hand and/or the free NH2 groups carried by the N-terminal amino acid or by amino acids inside the peptidic chain, for instance Lys, on the other hand, can be modified in so far as this modification does not alter the above mentioned properties of the polypeptide.
  • the polypeptides which are thus modified are naturally part of the invention.
  • the above mentioned carboxyl groups can be acylated or esterified. Other modifications are also part of the invention.
  • the amine or carboxyl functions or both of terminal amino acids can be themselves involved in the bond with other amino acids.
  • the N-terminal amino acid can be linked to the C-terminal amino acid of another peptide comprising from 1 to several amino acids.
  • any peptidic sequences resulting from the modification by substitution and/or by addition and/or by deletion of one or several amino acids of the polypeptides according to the invention are part of the invention in so far as this modification does not alter the above mentioned properties of said polypeptides.
  • the polypeptides according to the invention can be glycosylated or not, particularly in some of their glycosylation sites of the type Asn-X-Ser or Asn-X-Thr, X representing any amino acid.
  • An advantageous recombinant polypeptide included in the composition of the invention is SEQ ID NO: 6 since this polypeptide shows the highest frequency of antibody responses in CSF of MS patients with no reactivity in the control patients.
  • polypeptides are also possible depending on its intended use.
  • the polypeptide may be synthesized with an extra cysteine residue added. This extra cysteine residue is preferably added to the amino terminus and facilitates the coupling of the polypeptide to a carrier protein which is necessary to render the small polypeptide immunogenic.
  • the polypeptide is to be labeled for use in radioimmune assays, it may be advantageous to synthesize the protein with a tyrosine attached to either the amino or carboxyl terminus to facilitate iodination.
  • This polypeptide possesses therefore the primary sequence of the polypeptide above-mentioned but with additional amino acids which do not appear in the primary sequence of the protein and whose sole function is to confer the desired chemical properties to the polypeptide.
  • the invention provides for a kit to diagnose MS.
  • the following necessary or kit can be used, said necessary or kit comprising: a composition (comprising at least one polypeptide selected from SEQ ID NO: 1-8) according to the invention, or at least one fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8, reagents for making a medium appropriate for the immunological reaction to occur, reagents enabling to detect the antigen/antibody complex which has been produced by the immunological reaction, said reagents possibly having a label, or being liable to be recognized by a labeled reagent, more particularly in the case where the above mentioned polypeptide is not labeled.
  • a normalized cDNA library derived from active, chronic MS plaques, with varying degrees of demyelination and inflammatory activity which was originally cloned into the pT7T3-Pac vector, was cloned into the M13 filamentous phage display vectors pSPA, B and C.
  • These vectors allow expression of cDNA products (peptides) derived from MS brain plaques as a fusion to minor coat protein pVI of filamentous phage M13 in 3 reading frames for correct expression of the protein products.
  • a total library size of 1.1 x 10 7 colony forming units (cfu) was obtained.
  • MS cDNA display library For enrich the MS cDNA display library for cDNA products (displayed peptides) that are specifically bound by autoantibodies present in cerebrospinal fluid of MS patients, we performed successive rounds of selection (see Figure 3, and Table 7) on pooled CSF of 10 randomly selected RR MS patients. Following rescue of the phage clones after each of 4 rounds of selection, enriched phage clones, each bearing a single fusion peptide derived from the MS cDNA display library, were randomly selected for further study.
  • the results of the phage ELISA screening of the individual phage-cDNA clones on 167 different CSF are presented in Table 5. All antigens tested showed exclusive or preferential reactivity in the MS group as compared to the control group.
  • Clones UH-CSFP1.4-UH-CSFP1.7 showed reactivity in 17 of 73 (23%) MS CSF whereas no reactivity towards the OIND and NIND CSF specimens was observed.
  • the remaining clones (UH-CSFP1.1-UH-CSFP1.3 and UH-CSFP1.8) showed higher reactivity in the MS group 25/73 (34%) as compared to the control group 13/94 (14%), and therefore, these clones were also defined as clones with an MS-related serological profile.
  • 33 of 73 (45%) MS patients showed CSF IgG antibodies reactive with at least one of the panel of 8 antigenic targets.
  • the highest frequency of antibody responses in MS CSF with no reactivity in the control group was found to UH-CSFP1.6. All CSF samples tested showed equivalent total CSF IgG levels. CSF samples with high IgG concentration were normalized to the normal CSF concentration range.
  • UH-CSFP1.3 UH-CSFP1.6, UH-CSFP1.7 and UH-CSFP1.8
  • UH-CSFP1.7 was cloned as antigen (6,1 kDa)
  • the UH-CSFP1.8 (SPAG16) protein was cloned as partial (13,3 kDa, protein product as detected using SAS) and full-length protein (20,3 kDa) with a 16,7 kDa thioredoxin fusion (His tag) resulting in 22,8 kDa protein for UH-CSFP1.7 and 30 kDa partial and 37 kDa full length band for SPAG16 protein on SDS-PAGE after Coommassie staining (see Figure 2).
  • UH-CSFP1.3 Due to the presence of amber stop codons in the sequences of clones UH-CSFP1.3 and UH- CSFP1.6, site-directed mutagenesis was performed to create glutamine codon(s) for use in bacterial protein expression in the non-suppressing LMG194 strain. Following site-directed mutagenesis, UH-CSFP1.3 was cloned as antigen (6,1 1 kDa) resulting in a 22,8 kDa protein including thioredoxin (see Figure 6). Due to toxicity, the entire UH-CSFP1.6 could not be expressed. Therefore, the first part of the protein encoded by amino acids 1 -52 of the antigen (as detected using SAS) was produced, resulting in a 22,5 kDa protein product including thioredoxin (see Figure 6).
  • the produced monoclonal antibody showed the same epitope specificity as previously identified for MS serum or CSF samples. This allows further analysis of the UH-CSFP1.1 antigen.
  • the murine monoclonal antibody against UH-CSFP1.1 was used for immunohistochemical staining of experimental autoimmune encephalomyelitis (EAE) rat brain tissue. It was observed that the murine monoclonal antibody against UH-CSFP1.1 stained the endothelial lining of blood vessels and showed cytoplasmic staining of large neurons.
  • EAE autoimmune encephalomyelitis
  • Cerebrospinal fluid samples were obtained from 73 MS patients, 30 patients with other inflammatory (meningitis, polyneuropathy) and 64 patients with non-inflammatory neurological disorders (hernia, epilepsy, dementia, headache, Alzheimer patients, ...) undergoing lumbar puncture for diagnostic purposes. MS patients were diagnosed according to the McDonald and Poser criteria 14 . Characteristics of the study population are shown in table 1. From 28 out of 73 MS patients, paired serum samples were collected. CSF and serum samples were stored at -80 0 C after collection. The study was approved by the institutional ethics committee.
  • a normalized cDNA library (1.0 x 10 6 primary recombinants) derived from 3 active chronic MS plaques, with varying degrees of demyelination and inflammatory activity (gift from Dr. Soares) was used to construct an MS cDNA display library by cloning it as a fusion protein with filamentous phage minor protein pVI. Therefore, the library was transferred to our phage display vectors, named pSPVIA, pSPVIB and pSPVIC, each encoding one of three reading frames. Details of the cloning procedure are described in 15 .
  • the SAS procedure was performed as described previously (Somers, Jl) 15 .
  • CSF samples of 10 randomly selected untreated relapsing remitting (RR)-MS patients were pooled and used for affinity selections. Characteristics of the patients used for affinity selections are shown in table 2.
  • CSF samples were absorbed against Escherichia coli (E. coli) and phage antibodies as described in 15 .
  • E. coli Escherichia coli
  • phage antibodies as described in 15 .
  • pooled CSF was stored at -20 0 C.
  • pooled preabsorbed CSF was used for the selection procedure. Affinity selections were performed as described before 15 .
  • an immunotube (Nunc, Roskilde, Denmark) was coated with rabbit anti-human IgG (Dako, Glostrup, Denmark) in coating buffer (0.1 M sodium hydrogen carbonate pH 9.6) for 2 hours at 37 0 C. After washing the immunotube twice with phosphate-buffered saline containing 0.1% Tween 20 and twice with PBS, the tubes were blocked for 2 hours with 2% MPBS (2% milk powder in PBS). For the first round of the selection procedure, phage were prepared from the MS cDNA library cloned in the 3 phage display vectors pSP6A, B and C. Phage were prepared as described previously 16 .
  • phage were added to pooled preadsorbed CSF (1 :5 diluted in 4% MPBS) and incubated for 1.5 hour at RT on a rotating platform. After washing the coated immunotube twice with PBST and twice with PBS, the preincubated CSF and phage mix was transferred to the coated immunotube and incubated for 30 minutes on a rotating platform and 120 minutes standing at RT. Tubes were then washed extensively with PBST and PBS to remove non-binding phage. Binding phage were eluted with 100 mM triethylamine and neutralized with 1 M Tris HCI as described before 17 . E.
  • coli TG1 cells were infected with input and output phage and plated on 2 x TY agar plates containing ampicillin and glucose (16 g/l bacto-tryptone, 10 g/l yeast extract, 5 g/l NaCI, 15 g bacto-agar/l, ampicillin at 100 ⁇ g/ml and glucose at 2%) at each round of selection. Resultant colonies were scraped and phages were rescued for further rounds of affinity selections. To monitor enrichment of specific clones, input and output phage from each round of selection were titrated and the ratio of output/input phage was determined. After several rounds of selection, individual colonies were selected and the insert size and sequence was determined as described in 15 . Sequences were submitted to GenBank for BLAST homology search.
  • ELISA of ligand displaying phage was performed as described in 15 . Immunoreactivity for each phage peptide was measured in relation to an internal control signal detected by antibody reactivity against the empty phage.
  • CSF was pre-incubated in the presence of 0-50 pmol/50 ⁇ l synthetic peptide UH-CSFP1.1 (NH2-ASSRGYEDLRTF-COOH) or random peptide. Subsequently, the immunoreactivity to phage UH-CSFP1.1 was determined according to the standard phage ELISA procedure.
  • Plasmid was isolated using the Qiagen Plasmid Midi Kit according to the manufacturer's instructions.
  • the isolated plasmid was EcoR ⁇ /Not ⁇ digested and the excised DNA was gel- purified (GFXTM PCR DNA and Gel Band Purification Kit, GE Healthcare, Brussel, Belgium).
  • the excised DNA fragment was used as probe in Northern blot. Probes were labelled with [ ⁇ 32 P] using the High Prime DNA Labeling Kit (Roche, Vilvoorde, Belgium). Briefly, 50 ng excised DNA was first denatured during 10 min in boiling water and immediately chilled on ice. The labelling mix was added to the DNA and after 45 min incubation at 37°C, the reaction was stopped by addition of 0,2 M EDTA. Labelled DNA was purified with Sephadex G75 columns and radioactivity measured with a scintillation counter.
  • Northern blotting was performed using the Multiple Tissue Northern (MTNTM) Blot (BD Biosciences, Erembodegem, Belgium). Briefly, labelled DNA or human ⁇ -actin cDNA control probe was denatured at 97°C during 5 min and immediately chilled on ice for a few minutes. After prehybridisation of the blotting membrane with ExpressHyb solution, the radioactively labelled probe was added (2-10 ng/ml or 1-2 x 10 6 cpm/ml) and hybridization occurred overnight at 68 0 C. After washing 3 times, the blotting membrane was exposed to X-ray film at - 70 0 C and developed using the Gevamatic 60 (Agfa Gevaert, Mortsel, Belgium).
  • MTNTM Multiple Tissue Northern
  • UH-CSFP1.1 monoclonal antibody for UH-CSFP1.1 was produced according to the hybridoma technology developed by Kohler and Milstein (Kohler, G and Milstein, C, 1973, Nature 256, 495-497). Due to its small size, UH-CSFP1.1 peptide was coupled to keyhole limpit hemocyanin (KLH) as carrier (UH-CSFP1.1 ) (Eurogentec) for immunization of Balb/c female mice. After three intraperitoneal immunizations with 150 ⁇ g UH-CSFP1.1-KLH, spleen cells were isolated and fused with a mouse myeloma cell line (Sp2/0).
  • KLH keyhole limpit hemocyanin
  • ELISA on peptides For ELISA experiments, 96-well ELISA plates (Greiner) are coated with 100 ⁇ l of 1 ⁇ g/ml peptide (UH-CSFP1.1 , UH-CSFP1.2, UH-CSFP1.4 and UH-CSFP1.5) in PBS and kept overnight at RT. Wells are then washed with 3 times with PBS 0.05%Tween 20 and blocked at RT with blocking buffer (2% nonfat milk in PBS). After washing 3 times with PBS 0.05% Tween20, the plates are incubated with 100 ⁇ l diluted samples (CSF 1 :5 diluted and serum, 1 :100 diluted in blocking buffer) for 2 hours at RT.
  • CSF 1 :5 diluted and serum, 1 :100 diluted in blocking buffer 100 ⁇ l diluted samples
  • ELISA experiments were performed as described for peptides, except that 96-well ELISA plates (Greiner) were coated with 100 ⁇ l of 1 ⁇ g/ml purified proteins (UH-CSFP1.3, UH- CSFP1.6, UH-CSFP1.7 and UH-CSFP1.8) in coating buffer and kept overnight at 4°C. Serum samples were considered positive for antibodies against the purified proteins when the OD 450 was higher than the mean + 3 times the standard deviation of the healthy controls.
  • the horizontal line in figure 8 represents the cut-off value.
  • C NIND hernia, epilepsy, dementia, headache, migraine, Alzheimer, hydrocephalus d , OIND: meningitis, polyneuropathy

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The present invention relates to the diagnosis of neurological disorders, more specifically to the diagnosis of multiple sclerosis. A biomarker panel is provided which can be used to detect if a subject has multiple sclerosis.Also described are methods of identification of such biomarkers.

Description

Biomarkers for multiple sclerosis
Field of the invention
The present invention relates to the diagnosis of neurological disorders, more specifically to the diagnosis of multiple sclerosis. A biomarker panel is provided which can be used to detect if a subject has multiple sclerosis. Also described are methods of identification of such biomarkers.
Background to the invention Multiple sclerosis (MS) affects more than 350.000 people in the US and 2.5 million worldwide. In the US prevalence estimates vary between 5 and 119 per 100.000 and healthcare costs are estimated to be more than $10 billion annually in the US alone. It is the most common neurological disease in young adults, with the risk of subsequent chronic functional impairment and disability after 10-15% of disease duration. The disease is characterized initially in 80-90% of patients by recurrent neurological events (relapses) that are attributable to multifocal lesions within the CNS. Further disease courses vary from benign to classical relapsing-remitting (RR), primary (PP) and secondary (SP) chronic progressive or rare fulminant disease course. MS is considered to be of autoimmune origin and is characterized neuropathologically by variable extents of focal inflammation, demyelination, axonal damage, gliotic scarring and atrophy, but also by remyelination and regeneration in the CNS. This has led, together with the clinical variability, to the concept of MS as a heterogenous disease with respect to four pathogenetic mechanisms of demyelination 1'2. One of these pathogenetic subtypes is characterized neuropathologically by antibody-dependent immune mechanisms involved in the formation of MS lesions 1'3. During the past years, an important role of autoreactive B cells and autoantibodies has been demonstrated 4. Recent studies uniformly showed clonal expansion of antibody-secreting B cells in the CNS and cerebrospinal fluid (CSF) of patients with MS 5'6. Furthermore, detection of oligoclonal antibodies in CSF of patients with neurological diseases has been associated with the presence of MS. Numerous studies have reported the recognition of central nervous system (CNS) myelin autoantigens such as myelin basic protein (MBP), proteolipid lipoprotein, myelin oligodendrocyte glycoprotein, myelin associated glycoprotein by autoantibodies present in CSF and serum of MS patients, but also in patients with other-inflammatory neurological diseases (OIND) and non-inflammatory neurological diseases (NIND) as well as healthy controls 7"11. A physician may diagnose MS in some patients soon after the onset of the illness. In others, however, doctors may not be able to readily identify the cause of the symptoms, leading to years of uncertainty and multiple diagnoses. The vast majority of patients are mildly affected, but in the worst cases, MS can render a person unable to write, speak or walk. Unfortunately, no single laboratory test is yet available to prove or rule out MS. Therefore, there is a great need in the art for improved diagnostic tests for MS. The development of a panel of biomarkers, specific for different pathophysiological mechanisms, will be crucial for the further understanding of the pathogenesis of MS, as well as diagnosis, classification, disease activity, and theranostic applications.
In the present invention, we report the identification of autoantibody binding peptides/proteins which are highly specific for MS patients. The results obtained were also correlated to disease duration, disability and different clinical course of disease. The autoantibody profiles against these selected peptides can be used as a biomarker panel for the specific detection of MS.
Figures
Figure 1 : Expression profile of novel antigenic targets in normal tissues. Expression patterns are shown for UH-CSF1.4 and UH-CSFP1.7. The lower panel shows a control hybridization with an actin probe. Lane 1 : brain; lane 2: heart; lane 3: skeletal muscle; lane 4: colon; lane 5: thymus; lane 6: spleen; lane 7: kidney; lane 8: liver; lane 9: small intestine; lane 10: placenta; lane 1 1 : lung; lane 12: peripheral blood lymphocytes.
Figure 2: UH-CSFP1.7 and UH-CSFP1.8 protein expression in E.coli. UH-CSFP1.7 was cloned as antigen (6,1 kDa) and the UH-CSFP1.8 protein was cloned as partial* (13,3 kDa) and full-length protein (20,3 kDa) with a 16,7 kDa thioredoxin fusion (His tag) resulting in 22,8 kDa protein for UH-CSFP1.7 and 30 kDa partial and 37 kDa full length band for UH-CSFP1.8
(SPAG16 protein) on SDS-PAGE after Coommassie staining.
Figure 3: SAS procedure, a. A phage-displayed MS cDNA repertoire is preincubated with MS patient CSF antibodies, b. MS-specific antigens displayed on phage (black) bind to MS-antigen specific patient IgG (black), c. Phage antigen-lgG complexes (black) are captured on a surface coated with polyclonal anti-human IgG (checked), d. Nonrelevant phages are washed away, and CSF-IgG specific phages are eluted. e. Selected phages are used for reinfection of bacteria, f. Selected phages are amplified and used for further rounds of selection.
Figure 4: Solution phase assays demonstrate high affinity and specificity of CSF antibodies to UH-CSFP1.1. The UH-CSFP1.1 peptide was pre-incubated at different dilutions with MS-CSF8 and MS-CSF26, respectively, and subsequently, the remaining immunoreactivity measured by
ELISA. Competition by the UH-CSFP1.1 peptide is displayed. No competition was measured with the random peptide.
Figure 5: Histogram showing reactivity against UH-CSFP1.1 peptide of 10 random clones tested by ELISA assay. A positive signal was obtained for clone 7.
Figure 6: Protein expression of UH-CSFP1.3 and UH-CSFP1.6 (partial). Figure 7: Reactivity of 8 individual CSF samples against UH-CSFP1.1 , UH-CSFP1.2, UH- CSFP1.4, UH-CSFP1.5 and negative control.
Figure 8: Antibody reactivity towards UH-CSFP1.6 in serum from 16 randomly selected MS patients, 15 NIND/OIND patients and 16 healthy controls. The horizontal line represents the cut-off value.
Aims and detailed description of the invention
In the present invention we have identified a set of biomarkers which can be used for the detection of Multiple Sclerosis (MS) in patients. Biomarkers were isolated with the technology of Serological Antigen Selection (SAS) wherein antigens (i.e. biomarkers) were identified that bind to antibodies present in cerebrospinal fluid (CSF) in patients suffering from Multiple Sclerosis. More specifically, a cDNA phage display library comprising cDNA products derived from MS brain plaques - expressed as a fusion to minor coat protein pVI of filamentous phage M13 - was panned to identify cDNA clones that bind auto-antibodies in CSF specimens from MS patients. A biomarker panel of 8 antigenic cDNA targets which showed 86% specificity and 45% sensitivity in discriminating MS patients and controls was retrieved. Besides a role in the immediate (early) diagnosis of patients suspected for MS, the biomarker panel (i.e. the antigenic cDNA targets) can be used to assist in sub-typing MS patients. Thus in a first embodiment the invention provides a composition comprising at least two different polypeptides comprising a sequence represented by any of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8. Such a composition is herein also designated as a biomarker or as a biomarker panel. The SEQ ID NO: 1-8 correspond with the translated amino acid sequences of the antigens retrieved by the selection of phage displayed MS cDNA expression library on MS patient cerebrospinal fluid (CSF). Thus the translation of the insert of UH-CSFP1.1 corresponds with SEQ ID NO: 1 , ..., and the translation of the insert of UH-CSFP1.8 corresponds with SEQ ID NO: 8 (see table 3). The nucleotide sequences which encode SEQ ID NO: 1-8 are depicted in SEQ ID NO: 9-16 (wherein SEQ ID NO: 9 encodes SEQ ID NO: 1 , ..., and SEQ ID NO: 16 encodes SEQ ID NO: 8). Thus a composition comprises at least two different polypeptides wherein such a polypeptide comprises a sequence as depicted by SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7 or 8. This means that a polypeptide present in the composition can also be a protein. As an example SEQ ID NO:8 was cloned as a partial 13.3 kDa protein (protein product as detected using SAS). Since SEQ ID NO: 8 (corresponding with UH-CSFP1.8) is a fragment of the SPAG16 protein (which full length is 37 kDa) the composition can also comprise the full length SPAG16 protein. The composition of the invention can also comprise at least two different polypeptides wherein said polypeptides are fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7 or 8. It is envisaged that 5 consecutive amino acids derived from SEQ ID NO: 1-8 are sufficient to be recognized as antigens by the autoantibodies present in for example serum or CSF.
In a particular embodiment the composition comprises 8 different polypeptides comprising a sequence selected from SEQ ID NO: 1-8 or 8 different fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
In another particular embodiment the composition comprises 4 different polypeptides comprising a sequence represented by SEQ ID NO: 4, 5, 6 and 7 or 4 different fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 4, 5, 6 and 7. In another embodiment the invention provides the use of a composition of the invention for detecting the presence of specific antibodies to at least one polypeptide present in said composition wherein said antibodies are present in a body fluid of a mammal. In another particular embodiment the invention provides the use of a composition of the invention for detecting the presence of specific auto-antibodies to at least one polypeptide present in said composition wherein said auto-antibodies are present in a body fluid of a mammal. In particular embodiments said use of a composition is an "in vitro" use of a composition. The latter implies a diagnostic method with no direct interaction with the patient. The term 'body fluid' includes blood, blood serum, blood plasma, saliva, urine, tears, bone marrow fluid, cerebrospinal fluid (CSF), synovial fluid, lymphatic fluid, amniotic fluid, nipple aspiration fluid and the like. Preferred body fluids for analysis are those that are conveniently obtained from patients, particularly preferred body fluids include blood serum, blood plasma and CSF.
In yet another embodiment the invention provides a method for detecting multiple sclerosis in a mammal comprising i) detecting the presence of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the presence or quantity of said antibody indicates that said mammal suffers from multiple sclerosis. In yet another embodiment the method for detecting multiple sclerosis in a mammal of the present invention is combined with the detection of the MS markers described in US20040043431 and more specifically to the markers described in the claims 5, 6, 7, 8, 9 and 10 of said application.
In yet another embodiment the invention provides a method for evaluating the prognosis/disease severity of multiple sclerosis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the increased or decreased concentration of said antibody indicates the prognosis of multiple sclerosis in said mammal. In yet another embodiment the invention provides a method for selecting mammals for a specific therapeutic treatment of multiple sclerosis or evaluating the therapeutic treatment of multiple sclerosis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the increased or decreased concentration of said antibody leads to an election of a specific therapeutic treatment of multiple sclerosis in said mammal. In a preferred embodiment said body fluid is CSF. In yet another preferred embodiment said body fluid is serum. In another preferred embodiment said mammal is a human. In yet another embodiment the invention provides an antibody that specifically binds to a polypeptide selected from the group comprising of polypeptides selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8. Methods for generating antibodies are well known in the art. In a preferred embodiment the antibodies are monoclonal antibodies. For the purpose of generation of antibodies the polypeptides forming part of the compositions of the invention may be synthesized chemically or may be made in a recombinant way. They may also be coupled to a soluble carrier after synthesis or after recombinant production. If a carrier is used the nature of such a carrier should be such that it has a molecular weight greater than 5000 and should not be recognized by antibodies. Such a carrier can be a protein. Proteins which are frequently used as carriers are keyhole limpet hemocyanin, bovine gamma globulin, bovine serum albumin, and poly-L-lysine. There are many well described techniques for coupling peptides to carriers. The linkage may occur at the N-terminus, C-terminus or at an internal site in the peptide. The polypeptide may also be derivatized for coupling. The polypeptides may also be synthesized directly on an oligo-lysine core in which both the alpha as well as the epsilon-amino groups of lysines are used as growth points for the polypeptides. The number of lysines comprising the core is preferably 3 or 7. Additionally, a cysteine may be included near or at the C-terminus of the complex to facilitate the formation of homo- or heterodimers.
In general terms the invention relates to a process for detecting antibodies related to MS in a biological sample of a mammal liable to contain them, this process comprising contacting the biological sample with a composition according to the invention under conditions enabling an immunological reaction between said composition and the antibodies which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed. The detection can be carried out according to any classical process. By way of examples immunoenzymatic processes according to the ELISA technique or immunofluorescent or radioimmunological (RIA) or the equivalent ones (e.g. LINE blot or LINE assay) can be used. Thus the invention also relates to polypeptides according to the invention labeled by an appropriate label of the enzymatic, fluorescent, biotin, radioactive type. Such a method for detecting antibodies related to MS comprises for instance the following steps: deposit of determined amounts of a polypeptidic composition according to the invention on a support (e.g. into wells of a titration microplate), introduction on said support (e.g. into wells) of increasing dilutions of the body fluid (e.g. CSF) to be diagnosed, incubation of the support (e.g. microplate), repeated rinsing of the support (e.g. microplate), introduction on the support labeled antibodies which are specific for immunoglobulins present in the body fluid, the labeling of these antibodies being based on the activity of an enzyme which is selected from among the ones which are able to hydrolyze a substrate by modifying the absorption of the radiation of this latter at least at a given wave length, detection by comparing with a control standard of the amount of hydrolyzed substrate.
In yet another embodiment the invention also relates to a process for detecting and identifying antigens of MS in a body fluid liable to contain them, this process comprising: contacting the biological sample with an appropriate antibody of the invention (i.e. antibodies with a specificity for a polypeptide of the composition) under conditions enabling an immunological reaction between said antibody and the antigens of MS which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed.
Thus antibodies, in particular auto-antibodies, which recognize the polypeptides of the invention, can be detected in a variety of ways. One method of detection is further described in the examples and uses enzyme-linked immunosorbant assay (ELISA) of the polypeptides of the invention displayed by phages (i.e. phage-ELISA technology). The latter technology is fully described in Somers V. et al (2005) J. of Autoimmunity 25: 223-228, wherein paragraph 2.6 on page 225 is herein specifically incorporated). In other ways in the detection in ELISA a polypeptide or a mixture of polypeptides is bound to a solid support. In some cases, this will be a microtiter plate but may in principle be any sort of insoluble solid phase (e.g. glass, nitrocellulose). In one embodiment a suitable dilution or dilutions of for example CSF or serum to be tested is brought into contact with the solid phase to which the polypeptide is bound. In another embodiment "a solution hybridization" is carried out in which high affinity interactions occur (eg. biotinylated polypeptides of the composition are pre-incubated with CSF). The incubation is carried out for a time necessary to allow the binding reaction to occur.
Subsequently, unbound components are removed by washing the solid phase. The detection of immune complexes (i.e. auto-antibodies present in for example human CSF binding to at least one polypeptide of the invention) is achieved using antibodies which specifically bind to human immunoglobulins, and which have been labeled with an enzyme, preferably but not limited to either horseradish peroxidase, alkaline phosphatase, or beta-galactosidase, which is capable of converting a colorless or nearly colorless substrate or co-substrate into a highly colored product or a product capable of forming a colored complex with a chromogen. Alternatively, the detection system may employ an enzyme which, in the presence of the proper substrate(s), emits light. The amount of product formed is detected either visually, spectrophotometrically, electrochemically, fluorescently or luminometrically, and is compared to a similarly treated control. The detection system may also employ radioactively labeled antibodies, in which case the amount of immune complex is quantified by scintillation counting or gamma counting. Other detection systems which may be used include those based on the use of protein A derived from Staphylococcus aureus Cowan strain I, protein G from group C Staphylococcus sp. (strain 26RP66), or systems which make use of the high affinity biotin- avidin or streptavidin binding reaction.
The polypeptides of the invention may be either labeled or unlabeled. Labels which may be employed may be of any type, such as enzymatic, chemical, fluorescent, luminescent, or radioactive. In addition, the polypeptides may be modified for binding to surfaces or solid phases, such as, for example, microtiter plates, nylon membranes, glass or plastic beads, and chromatographic supports such as cellulose, silica, or agarose. The methods by which polypeptides can be attached or bound to solid support or surface are well known to those skilled in the art.
The polypeptides of the invention can be prepared according to the classical techniques in the field of peptide synthesis. The synthesis can be carried out in homogeneous solution or in solid phase. For instance, the synthesis technique in homogeneous solution which can be used is the one described by Houbenweyl in the book titled "Methode der organischen chemie"
(Method of organic chemistry) edited by E. Wunsh, vol. 15-1 et II. THIEME, Stuttgart 1974. The polypeptides of the invention can also be prepared in solid phase according to the method described by Atherton & Shepard in their book titled "Solid phase peptide synthesis" (Ed. IRL
Press, Oxford, NY, Tokyo, 1989). Synthesis protocols in the art generally employ the use of t- butyloxycarbonyl- or 9-fluorenylmethoxy-carbonyl-protected activated amino acids. The procedures for carrying out the syntheses, the types of side-chain protection, and the cleavage methods are amply described in, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2nd Edition, Pierce Chemical Company, 1984; and Atherton and Sheppard, Solid
Phase Peptide Synthesis, IRL Press, 1989. In yet another embodiment antibodies raised to polypeptides of the invention (or carrier-bound polypeptides) can also be used in conjunction with labeled polypeptides of the invention for the detection of (auto)-antibodies present in serum or CSF by competition assay. In this case, antibodies raised to polypeptides are attached to a solid support which may be, for example, a plastic bead or a plastic tube. Labeled polypeptide is then mixed with suitable dilutions of the fluid (e.g. CSF) to be tested and this mixture is subsequently brought into contact with the antibody bound to the solid support. After a suitable incubation period, the solid support is washed and the amount of labeled polypeptide is quantified. A reduction in the amount of label bound to the solid support is indicative of the presence of (auto)-antibodies in the original sample. By the same token, the polypeptide may also be bound to the solid support. Labeled antibody may then be allowed to compete with (auto)-antibody present in the sample (e.g. CSF) under conditions in which the amount of polypeptide is limiting. As in the previous example, a reduction in the measured signal is indicative of the presence of (auto)-antibodies in the sample tested.
In a particular embodiment a test for giving evidence of the fact that one or more polypeptides present in a composition of the invention are recognized by antibodies present in for example CSF of serum (for example auto-antibodies present in CSF of multiple sclerosis patients) is an immunoblotting (or Western blotting) analysis. In the latter case polypeptides can be chemically synthesized or polypeptides (or the protein) can be produced via recombinant techniques. In short, after sodium dodecyl sulfate-polyacrylamide gel electrophoresis, polypeptides of the invention are blotted onto nitrocellulose membranes (e.g. Hybond C. (Amersham)) as described by Towbin H. et al., 1979, "Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications", Proc. Natl. Acad. Sci. USA 76:4350-4354. In order to identify selective recognition of polypeptides (or proteins) of the invention by CSF, nitrocellulose sheets are incubated overnight with each of these samples (e.g. diluted 1 :50) (after blocking a-specific protein-binding sites). Reactive areas on the nitrocellulose sheets are revealed by incubation with e.g. peroxidase conjugated goat anti-human immunoglobulin G antibody (e.g. diluted 1 :200) for 4 h, and after repeated washings, color reaction is developed by adding for example alpha-chloronaphtol (Bio-Rad Laboratories, Richmond, Calif.) in the presence of hydrogen peroxide.
It goes without saying that the free reactive functions which are present in some of the amino acids, which are part of the constitution of the polypeptides of the invention, particularly the free carboxyl groups which are carried by the groups GIu and Asp or by the C-terminal amino acid on the one hand and/or the free NH2 groups carried by the N-terminal amino acid or by amino acids inside the peptidic chain, for instance Lys, on the other hand, can be modified in so far as this modification does not alter the above mentioned properties of the polypeptide. The polypeptides which are thus modified are naturally part of the invention. The above mentioned carboxyl groups can be acylated or esterified. Other modifications are also part of the invention. Particularly, the amine or carboxyl functions or both of terminal amino acids can be themselves involved in the bond with other amino acids. For instance, the N-terminal amino acid can be linked to the C-terminal amino acid of another peptide comprising from 1 to several amino acids.
Furthermore, any peptidic sequences resulting from the modification by substitution and/or by addition and/or by deletion of one or several amino acids of the polypeptides according to the invention are part of the invention in so far as this modification does not alter the above mentioned properties of said polypeptides. The polypeptides according to the invention can be glycosylated or not, particularly in some of their glycosylation sites of the type Asn-X-Ser or Asn-X-Thr, X representing any amino acid.
An advantageous recombinant polypeptide included in the composition of the invention is SEQ ID NO: 6 since this polypeptide shows the highest frequency of antibody responses in CSF of MS patients with no reactivity in the control patients.
Variations of these polypeptides are also possible depending on its intended use. For example, if the polypeptide is to be used to raise antisera, the polypeptide may be synthesized with an extra cysteine residue added. This extra cysteine residue is preferably added to the amino terminus and facilitates the coupling of the polypeptide to a carrier protein which is necessary to render the small polypeptide immunogenic. If the polypeptide is to be labeled for use in radioimmune assays, it may be advantageous to synthesize the protein with a tyrosine attached to either the amino or carboxyl terminus to facilitate iodination. This polypeptide possesses therefore the primary sequence of the polypeptide above-mentioned but with additional amino acids which do not appear in the primary sequence of the protein and whose sole function is to confer the desired chemical properties to the polypeptide.
In yet another embodiment the invention provides for a kit to diagnose MS. To carry out the diagnostic method for MS, the following necessary or kit can be used, said necessary or kit comprising: a composition (comprising at least one polypeptide selected from SEQ ID NO: 1-8) according to the invention, or at least one fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8, reagents for making a medium appropriate for the immunological reaction to occur, reagents enabling to detect the antigen/antibody complex which has been produced by the immunological reaction, said reagents possibly having a label, or being liable to be recognized by a labeled reagent, more particularly in the case where the above mentioned polypeptide is not labeled.
Other characteristics and advantages of the invention will appear in the following examples and the figures illustrating the invention.
Examples
1. Enrichment of a phage displayed MS cDNA library with MS CSF
To create an MS cDNA display library, a normalized cDNA library derived from active, chronic MS plaques, with varying degrees of demyelination and inflammatory activity, which was originally cloned into the pT7T3-Pac vector, was cloned into the M13 filamentous phage display vectors pSPA, B and C. These vectors allow expression of cDNA products (peptides) derived from MS brain plaques as a fusion to minor coat protein pVI of filamentous phage M13 in 3 reading frames for correct expression of the protein products. A total library size of 1.1 x 107 colony forming units (cfu) was obtained.
To enrich the MS cDNA display library for cDNA products (displayed peptides) that are specifically bound by autoantibodies present in cerebrospinal fluid of MS patients, we performed successive rounds of selection (see Figure 3, and Table 7) on pooled CSF of 10 randomly selected RR MS patients. Following rescue of the phage clones after each of 4 rounds of selection, enriched phage clones, each bearing a single fusion peptide derived from the MS cDNA display library, were randomly selected for further study.
2. Characterization of the enriched phage clones
Among the enriched clones, a total of 52 clones were selected. cDNA inserts were sequenced and the translated protein sequences were determined. Sequence analysis revealed 8 antigenic targets, which we annotated with the name UH-CSFP-number, which is short for
University Hasselt - cerebrospinal fluid pool - number of the clone. These sequences corresponded to known proteins expressed in the correct reading frame, but also homology to untranslated regions of expressed genes, such as 3' UTR sequence of proteolipid protein, or homology to out of frame sequences were obtained (see table 3).
In initial experiments, we assessed the reactivity of the individual MS CSF specimens used for the selection procedure against 8 enriched antigenic cDNAs. As shown in Table 4, of the 10 CSF samples from RR-MS patients, 8 contained antibodies that reacted with at least 2 phage- peptide clones. These clones were used for subsequent screening on a large panel of CSF from other MS patients as well as CSF from patients with other inflammatory (OIND) and noninflammatory neurological disorders (NIND). Of the 10 RR-MS patients used for the selection procedure, paired serum samples were also collected and used for screening for antibody reactivity towards the 8 enriched antigenic cDNAs. In 3 out of 8 patients with antigen specific antibodies present in CSF, reactivity towards 1 of the 8 antigenic cDNAs was also found in paired serum. There was a good association between the positive signal observed in the CSF, and the reactivity demonstrated in serum. The signal on individual CSF tested was higher than that on individual serum tested, which is consistent with the dilution of the antigen specific antibodies present in the serum, when antibodies are intrathecal^ produced. When low reactivity in the CSF was observed, no positive signal was found in the serum of the same patient. In addition, no reactivity in paired serum samples was demonstrated in patients with antibody negative CSF (data not shown).
3. Detailed serological analysis of the MS panel
Next, clones were tested on a large panel of individual CSF specimens not used for the selection procedure (n= 63 for MS patients (54 RR-MS, 3 SP-MS patients and 6 PP-MS), n= 30 for OIND patients and n= 64 for NIND patients). The results of the phage ELISA screening of the individual phage-cDNA clones on 167 different CSF are presented in Table 5. All antigens tested showed exclusive or preferential reactivity in the MS group as compared to the control group. Clones UH-CSFP1.4-UH-CSFP1.7 showed reactivity in 17 of 73 (23%) MS CSF whereas no reactivity towards the OIND and NIND CSF specimens was observed. The remaining clones (UH-CSFP1.1-UH-CSFP1.3 and UH-CSFP1.8) showed higher reactivity in the MS group 25/73 (34%) as compared to the control group 13/94 (14%), and therefore, these clones were also defined as clones with an MS-related serological profile. In total, 33 of 73 (45%) MS patients showed CSF IgG antibodies reactive with at least one of the panel of 8 antigenic targets. The highest frequency of antibody responses in MS CSF with no reactivity in the control group was found to UH-CSFP1.6. All CSF samples tested showed equivalent total CSF IgG levels. CSF samples with high IgG concentration were normalized to the normal CSF concentration range.
4. Expression pattern of novel MS markers Northern blot analysis of the antigenic targets with no reactivity in the control group was performed on a variety of normal human tissues. UH-CSFP1.4 gave a transcript of 1.9 kb and was highly expressed brain, heart and placenta, and to a lower extent in skeletal muscle, kidney and liver. UH-CSFP1.7 gave a transcript size of 5,1 kb and showed a high expression in brain, heart and skeletal muscle. No transcript could be detected for UH-CSFP1.5 and UH- CSFP1.6.
We further selected 4 of the antigenic targets (UH-CSFP1.3, UH-CSFP1.6, UH-CSFP1.7 and UH-CSFP1.8), for protein expression in E.coli. UH-CSFP1.7 was cloned as antigen (6,1 kDa) and the UH-CSFP1.8 (SPAG16) protein was cloned as partial (13,3 kDa, protein product as detected using SAS) and full-length protein (20,3 kDa) with a 16,7 kDa thioredoxin fusion (His tag) resulting in 22,8 kDa protein for UH-CSFP1.7 and 30 kDa partial and 37 kDa full length band for SPAG16 protein on SDS-PAGE after Coommassie staining (see Figure 2). Due to the presence of amber stop codons in the sequences of clones UH-CSFP1.3 and UH- CSFP1.6, site-directed mutagenesis was performed to create glutamine codon(s) for use in bacterial protein expression in the non-suppressing LMG194 strain. Following site-directed mutagenesis, UH-CSFP1.3 was cloned as antigen (6,1 1 kDa) resulting in a 22,8 kDa protein including thioredoxin (see Figure 6). Due to toxicity, the entire UH-CSFP1.6 could not be expressed. Therefore, the first part of the protein encoded by amino acids 1 -52 of the antigen (as detected using SAS) was produced, resulting in a 22,5 kDa protein product including thioredoxin (see Figure 6).
5. Autoantibody reactivity and clinical data We next determined whether reactivity to our antigenic panel was associated with a particular disease phenotype. Autoantibody reactivity to at least 1 of the 8 antigenic targets was demonstrated in 30/64 (47%) RR-MS patients, 3/6 (50%) PP-MS patients and 0/3 SP-MS patients. Demographic variables and EDSS score in antibody-positive and antibody-negative MS patients are shown in Table 6. No differences were observed in age between antibody- positive and antibody-negative patients. Antibody reactivity could be observed in some patients at time of diagnosis and was present in patients with short disease duration (<1 year), but also in patients with a disease duration greater than 10 years. However, no correlation was found between antibody reactivity and disease duration. In order to assess the influence of antibody reactivity on disease severity, we examined the relationship between antibody reactivity and EDSS score. Antibody reactivity was found in 21/50 (42%) of patients with EDSS <3, 6/1 1 (54%) of patients with EDSS=3 or 3,5, and 3/5 (60%) of MS patients with EDSS=4. Although a higher percentage of patients showed reactivity to the panel of 8 antigenic cDNAs with increasing EDSS score, this difference was not significant.
6. Solution phase assay/Competition ELISA
To determine whether the observed autoantibody signature of MS CSF is due to the MS brain plaque derived peptides, 2 MS CSF specimens (one positive (MS-CSF8) and one negative (MS-CSF26) for UH-CSFP1.1 were pre-incubated with the synthetic peptide UH-CSFP1.1 (NH2-ASSRGYEDLRTF-COOH) representing the cDNA insert of clone UH-CSFP1.1 and with a non-specific (random) peptide. As shown in figure 4, preincubation with UH-CSFP1.1 peptide clearly inhibited the formation of specific IgG antibody/phage UH-CSFP1.1 complexes for MS- CSF8 while no inhibition was found for MS-CSF26. In contrast, CSF reactivity against clone UH-CSFP1.1 was not inhibited by addition of the random peptide.
7. Monoclonal antibody production A murine monoclonal antibody for UH-CSFP1.1 was produced based on the hybridoma technology developed by Kohler and Milstein (Kohler, G and Milstein, C, 1973, Nature 256, 495-497). Figure 5 represents antibody reactivity against UH-CSFP1.1 peptide following hybridoma selection. The ODs of supernatants from 10 random tested clones of hybridomas are indicated at first screening for antibody production. A positive ELISA signal was obtained for clone 7. Further subcloning of this clone resulted in a monoclonal hybridoma cell line producing antibodies directed against UH-CSFP1.1 peptide. The produced monoclonal antibody showed the same epitope specificity as previously identified for MS serum or CSF samples. This allows further analysis of the UH-CSFP1.1 antigen. In an alternative approach, we are using phage particles expressing the UH-CSFP antigenic targets for immunization of Balb/c female mice. Advantages of using phage-displayed peptides is that they are cheap, easy to obtain and that the antigen is displayed to the murine immune system as it is recognized in serum or CSF from MS patients.
8. ELISA on peptides To address whether antibody reactivity was also observed against linear peptides, we used ELISA on synthetic peptides (UH-CSFP1.1 , UH-CSFP1.2, UH-CSFP1.4 and UH-CSFP1.5). As shown in figure 7, MS patient No. 4 showed CSF reactivity against clone UH-CSFP1.1 , while for the other peptides, no reactivity was found. For the other MS patients, no reactivity was seen against any of the tested peptides. These results were consistent with the phage ELISA results for UH-CSFP1.1 , UH-CSFP1.2, UH-CSFP1.4 and UH-CSFP1.5.
9. ELISA on purified recombinant proteins
After protein expression of UH-CSFP1.3, UH-CSFP1.6, UH-CSFP1.7 and UH-CSFP1.8 (as described in example 4, second paragraph), immunoreactivity for each purified recombinant protein was measured in serum. Figure 8 represents antibody reactivity towards UH-CSFP1.6 in serum from 16 randomly selected MS patients, 15 NIND/OIND patients and 16 healthy controls. Reactivity was demonstrated in 3/16 MS patients and 1/15 NIND/OIND patients, while no reactivity was found in healthy controls.
10. lmmunohistochemical staining
After monoclonal antibody production (example 7), the murine monoclonal antibody against UH-CSFP1.1 was used for immunohistochemical staining of experimental autoimmune encephalomyelitis (EAE) rat brain tissue. It was observed that the murine monoclonal antibody against UH-CSFP1.1 stained the endothelial lining of blood vessels and showed cytoplasmic staining of large neurons.
Materials and methods 1. Patients and controls
Cerebrospinal fluid samples were obtained from 73 MS patients, 30 patients with other inflammatory (meningitis, polyneuropathy) and 64 patients with non-inflammatory neurological disorders (hernia, epilepsy, dementia, headache, Alzheimer patients, ...) undergoing lumbar puncture for diagnostic purposes. MS patients were diagnosed according to the McDonald and Poser criteria 14. Characteristics of the study population are shown in table 1. From 28 out of 73 MS patients, paired serum samples were collected. CSF and serum samples were stored at -800C after collection. The study was approved by the institutional ethics committee.
2. Cloning of an MS cDNA library for pVI display and Serological antigen selection (SAS) of phage pVI displayed cDNA repertoires
A normalized cDNA library (1.0 x 106 primary recombinants) derived from 3 active chronic MS plaques, with varying degrees of demyelination and inflammatory activity (gift from Dr. Soares) was used to construct an MS cDNA display library by cloning it as a fusion protein with filamentous phage minor protein pVI. Therefore, the library was transferred to our phage display vectors, named pSPVIA, pSPVIB and pSPVIC, each encoding one of three reading frames. Details of the cloning procedure are described in 15.
The SAS procedure was performed as described previously (Somers, Jl) 15. In brief, CSF samples of 10 randomly selected untreated relapsing remitting (RR)-MS patients were pooled and used for affinity selections. Characteristics of the patients used for affinity selections are shown in table 2. Before the start of the selection procedure, CSF samples were absorbed against Escherichia coli (E. coli) and phage antibodies as described in 15. Following adsorption, pooled CSF was stored at -200C. Subsequently, pooled preabsorbed CSF was used for the selection procedure. Affinity selections were performed as described before 15. In brief, an immunotube (Nunc, Roskilde, Denmark) was coated with rabbit anti-human IgG (Dako, Glostrup, Denmark) in coating buffer (0.1 M sodium hydrogen carbonate pH 9.6) for 2 hours at 370C. After washing the immunotube twice with phosphate-buffered saline containing 0.1% Tween 20 and twice with PBS, the tubes were blocked for 2 hours with 2% MPBS (2% milk powder in PBS). For the first round of the selection procedure, phage were prepared from the MS cDNA library cloned in the 3 phage display vectors pSP6A, B and C. Phage were prepared as described previously 16. Approximately 1013 phage were added to pooled preadsorbed CSF (1 :5 diluted in 4% MPBS) and incubated for 1.5 hour at RT on a rotating platform. After washing the coated immunotube twice with PBST and twice with PBS, the preincubated CSF and phage mix was transferred to the coated immunotube and incubated for 30 minutes on a rotating platform and 120 minutes standing at RT. Tubes were then washed extensively with PBST and PBS to remove non-binding phage. Binding phage were eluted with 100 mM triethylamine and neutralized with 1 M Tris HCI as described before 17. E. coli TG1 cells were infected with input and output phage and plated on 2 x TY agar plates containing ampicillin and glucose (16 g/l bacto-tryptone, 10 g/l yeast extract, 5 g/l NaCI, 15 g bacto-agar/l, ampicillin at 100 μg/ml and glucose at 2%) at each round of selection. Resultant colonies were scraped and phages were rescued for further rounds of affinity selections. To monitor enrichment of specific clones, input and output phage from each round of selection were titrated and the ratio of output/input phage was determined. After several rounds of selection, individual colonies were selected and the insert size and sequence was determined as described in 15. Sequences were submitted to GenBank for BLAST homology search.
3. Phage ELISA
ELISA of ligand displaying phage was performed as described in 15. Immunoreactivity for each phage peptide was measured in relation to an internal control signal detected by antibody reactivity against the empty phage. For competition ELISA, CSF was pre-incubated in the presence of 0-50 pmol/50 μl synthetic peptide UH-CSFP1.1 (NH2-ASSRGYEDLRTF-COOH) or random peptide. Subsequently, the immunoreactivity to phage UH-CSFP1.1 was determined according to the standard phage ELISA procedure.
4. Northern blot analysis
Plasmid was isolated using the Qiagen Plasmid Midi Kit according to the manufacturer's instructions. The isolated plasmid was EcoR\/Not\ digested and the excised DNA was gel- purified (GFX™ PCR DNA and Gel Band Purification Kit, GE Healthcare, Brussel, Belgium). The excised DNA fragment was used as probe in Northern blot. Probes were labelled with [α32P] using the High Prime DNA Labeling Kit (Roche, Vilvoorde, Belgium). Briefly, 50 ng excised DNA was first denatured during 10 min in boiling water and immediately chilled on ice. The labelling mix was added to the DNA and after 45 min incubation at 37°C, the reaction was stopped by addition of 0,2 M EDTA. Labelled DNA was purified with Sephadex G75 columns and radioactivity measured with a scintillation counter.
Northern blotting was performed using the Multiple Tissue Northern (MTN™) Blot (BD Biosciences, Erembodegem, Belgium). Briefly, labelled DNA or human β-actin cDNA control probe was denatured at 97°C during 5 min and immediately chilled on ice for a few minutes. After prehybridisation of the blotting membrane with ExpressHyb solution, the radioactively labelled probe was added (2-10 ng/ml or 1-2 x 106 cpm/ml) and hybridization occurred overnight at 680C. After washing 3 times, the blotting membrane was exposed to X-ray film at - 700C and developed using the Gevamatic 60 (Agfa Gevaert, Mortsel, Belgium).
5. Cloning of antigenic cDNAs in pBAD/Thio-TOPO vector and expression of recombinant proteins
Several of the antigenic cDNAs were cloned into the pBAD/Thio-TOPO vector (Invitrogen Life Technologies, Merelbeke, Belgium) and transformed into LMG194 cells according to the manuacturer's directions. Clones were cultured in LB Broth Base medium (Invitrogen Life Technologies, Merelbeke, Belgium) supplemented with ampicillin. Expression in E.coli, driven by the araBAD promoter (PBAD), was induced by addition of 0.2% arabinose. Recombinant proteins were expressed as fusions to His-Patch thioredoxin and were purified by Ni-NTA beads (Qiagen, Venlo, the Netherlands) according to the manufacturer's instructions. Expression of the proteins of the correct size was confirmed by SDS-PAGE. Protein identity was confirmed by mass spectrometry. Due to the presence of amber stop codons in the nucleotide sequences of clones UH-CSFP1.3 and UH-CSFP1.6, site-directed mutagenesis (Quikchange Site-Directed Mutagenesis Kit, Stratagene) was performed according to the manufacturer's directions in order to create glutamine codon(s) for use in bacterial protein expression in the non-suppressing LMG194 strain. For UH-CSFP1.6 the first part of the protein encoded by amino acids 1-52 of the antigen (as detected using SAS) was produced.
6. Statistical analysis
Statistical analysis was performed using GraphPad Prism version 4.0. Quantitative demographic variables for antibody-positive and antibody-negative individuals were compared using t-tests, and categorical variables were compared using chi-square tests. A p value <0.05 was considered statistically significant. Correlations between various markers were determined by linear regression analysis.
7. Monoclonal antibody production A murine monoclonal antibody for UH-CSFP1.1 was produced according to the hybridoma technology developed by Kohler and Milstein (Kohler, G and Milstein, C, 1973, Nature 256, 495-497). Due to its small size, UH-CSFP1.1 peptide was coupled to keyhole limpit hemocyanin (KLH) as carrier (UH-CSFP1.1 ) (Eurogentec) for immunization of Balb/c female mice. After three intraperitoneal immunizations with 150 μg UH-CSFP1.1-KLH, spleen cells were isolated and fused with a mouse myeloma cell line (Sp2/0). After selection of fused hybridomas by culturing in HAT medium, screening of the resulting hybridoma cell lines was performed by peptide ELISA using coated UH-CSFP1.1 peptide (Eurogentec) and cell line supernatant. After subcloning, a monoclonal hybridoma cell line was obtained, which produced antibodies directed against UH-CSFP1.1 peptide.
8. ELISA on peptides For ELISA experiments, 96-well ELISA plates (Greiner) are coated with 100 μl of 1 μg/ml peptide (UH-CSFP1.1 , UH-CSFP1.2, UH-CSFP1.4 and UH-CSFP1.5) in PBS and kept overnight at RT. Wells are then washed with 3 times with PBS 0.05%Tween 20 and blocked at RT with blocking buffer (2% nonfat milk in PBS). After washing 3 times with PBS 0.05% Tween20, the plates are incubated with 100 μl diluted samples (CSF 1 :5 diluted and serum, 1 :100 diluted in blocking buffer) for 2 hours at RT. After several washings with PBS-T, wells are incubated with 100 μl of 1 :2000 dilution of HRP-conjugated anti-human IgG in blocking buffer for 1 hour. After washing, 100 μl TMB-developing solution is added to each well, which is then incubated at RT. The reaction is stopped by the addition of 1 M H2SO4 and read at 450 nm. For negative controls, wells are not incubated with sample, and other wells are not coated with antigen but are incubated with sample. For negative control values, the mean of both negative control values are presented.
9. ELISA on purified recombinant proteins
ELISA experiments were performed as described for peptides, except that 96-well ELISA plates (Greiner) were coated with 100 μl of 1 μg/ml purified proteins (UH-CSFP1.3, UH- CSFP1.6, UH-CSFP1.7 and UH-CSFP1.8) in coating buffer and kept overnight at 4°C. Serum samples were considered positive for antibodies against the purified proteins when the OD450 was higher than the mean + 3 times the standard deviation of the healthy controls. The horizontal line in figure 8 represents the cut-off value.
Table 1. Characteristics of the study population
Mean age (SD)
Diagnosis No. Female/Male (range) in years
MS 73 51/22 38,6 (9,5) (16-57)
RR-MS 64 37,4 (9,0) (16-56)
SP-MS 3 48,7 (1 1 ) (36-56)
PP-MS 6 47,3 (8,3) (38-57)
NIND 64 34/30 55,4 (17,6) (21-93)
OIND 30 15/15 43,5 (15) (19-81 ) Table 2. Characteristics of patients used for affinity selections
Subject Gender Age Age start Disease Diagnosis EDSS
(M/F) (years) disease duration (years) (years)
1 49 40 8 RR-MS 3,5
M 42 34 8 RR-MS 1,5
52 49 3 RR-MS 1,5
41 38 3 RR-MS 3,0
52 51 0.5 RR-MS 1,0
48 46 0.6 RR-MS 1,5
46 44 1 RR-MS 2,5
43 35 7 RR-MS 1,5
36 32 3 RR-MS 1,5
10 M 31 27 4 RR-MS 0
Table 3. Sequence analysis of antigens retrieved by the selection of phage displayed MS cDNA expression library on MS patient CSF
Antigens
Vector Translated amino
Name GenBank No. pSPa Identity acid sequenceb Sizec Comments
UH-CSFP1.1 NM014382 B HS ATP2C1 , transcript variant 1 ASSRGYEDLR 12 3' UTR
UH-CSFP1.2 NM199478 B PLP1 , transcript variant 2 LDNSYHDNPV 23 3' UTR
UH-CSFP1.3 BX509701.1 A HS DKFZp686A1481 LRAPAGLGAA 52 Est
UH-CSFP1.4 BC006427 C HS KIAA1279 GARCINAEQP 14 out of frame
UH-CSFP1.5 NM00729 B HS PACSI N2 YSCLKLYSFA 1 1 3' UTR
UH-CSFP1.6 AC114947.2 A HS chromosome 5 clone CTD-2636A23 EHATQNQVSV 103
UH-CSFP1.7 BC032450 A HS chromosome 10 ORF with retained intron GTGSGQGEEA 54
UH-CSFP1.8 BC067756.1 A HS sperm associated antigen 16 ADDNFSIPEG 121 in-frame
a Reading frame of vector b Translated amino acid sequence of the cDNA insert according to the reading frame of the vector: the first 10 aa of the fusion product are presented c Size of protein product in amino acids fused to pVI coat protein, * stop codon
Table 4. Reactivity of panel of 8 phage clones on individual MS CSF used for the selection procedure
Figure imgf000021_0001
positive ELISA signal at OD450 nm (> 1.5x background) negative ELISA signal at OD450 nm (< 1.5x background) Table 5. ELISA screening of individual phage-cDNA clones on 167 different CSF
Non- Total Total
Name Selection Selection NINDC OINDd Controls a b
UH-CSFP1.1 3/10 3/63 6/73 3/64 1/30 4/94
UH-CSFP1.2 4/10 6/63 10/73 1/64 0/30 1/94
UH-CSFP1.3 1/10 4/63 5/73 0/64 1/30 1/94
UH-CSFP1.4 3/10 1/63 4/73 0/64 0/30 0/94
UH-CSFP1.5 4/10 2/63 6/73 0/64 0/30 0/94
UH-CSFP1.6 6/10 4/63 10/73 0/64 0/30 0/94
UH-CSFP1.7 1/10 4/63 5/73 0/64 0/30 0/94
UH-CSFP1.8 1/10 13/63 14/73 7/64 0/30 7/94
a individual antigen reactive CSF from MS patients used in the selection procedure b individual CSF from MS patients not used in the selection procedure
C NIND: hernia, epilepsy, dementia, headache, migraine, Alzheimer, hydrocephalus d , OIND: meningitis, polyneuropathy
Table 6. Comparison of antibody-positive and antibody-negative patients with established MS
Antibody Antibody
Positive Negative
Characteristic (n=33) (n=40) P
Age, mean ± SD years 37,7 ± 8.9 39,4 ± 10,0 NS
Disease duration, mean ± SD years 33,,66 ±± 33..33 4,3 ± 5,2 NS
Sex
Male 1 1 1 1
Female 22 29 EDSS, gem ± SD 2 ± 1 2 ± 1 NS
Age and disease duration were compared by Mest, and categorical variables were compared by chi-square testing with appropriate degrees of freedom. NS= not significant
References
1. Lucchinetti, C. F., Bruck, W., Rodriguez, M., and Lassmann, H. Distinct patterns of multiple sclerosis pathology indicates heterogeneity on pathogenesis. Brain Pathol., 6: 259-274, 1996.
2. Lucchinetti, C, Bruck, W., Parisi, J., Scheithauer, B., Rodriguez, M., and Lassmann, H.
Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann. Neurol., 47: 707-717, 2000.
3. Genain, C. P., Cannella, B., Hauser, S. L., and Raine, C. S. Identification of autoantibodies associated with myelin damage in multiple sclerosis. Nat. Med., 5: 170- 175, 1999.
4. Cross, A. H., Trotter, J. L., and Lyons, J. B cells and antibodies in CNS demyelinating disease. J.Neuroimmunol., 112: 1-14, 2001.
5. Qin, Y., Duquette, P., Zhang, Y., Talbot, P., Poole, R., and Antel, J. Clonal expansion and somatic hypermutation of V(H) genes of B cells from cerebrospinal fluid in multiple sclerosis. J. Clin. Invest, 102: 1045-1050, 1998.
6. Baranzini, S. E., Jeong, M. C, Butunoi, C, Murray, R. S., Bernard, C. C, and Oksenberg, J. R. B cell repertoire diversity and clonal expansion in multiple sclerosis brain lesions. J.Immunol., 163: 5133-5144, 1999.
7. Kami, A., Bakimer-Kleiner, R., Abramsky, O., and Ben Nun, A. Elevated levels of antibody to myelin oligodendrocyte glycoprotein is not specific for patients with multiple sclerosis. Arch. Neurol., 56: 311-315, 1999.
8. Moller, J. R., Johnson, D., Brady, R. O., Tourtellotte, W. W., and Quarles, R. H. Antibodies to myelin-associated glycoprotein (MAG) in the cerebrospinal fluid of multiple sclerosis patients. J.Neuroimmunol., 22: 55-61 , 1989.
9. Vojdani, A., Vojdani, E., and Cooper, E. Antibodies to myelin basic protein, myelin oligodendrocytes peptides, alpha-beta-crystallin, lymphocyte activation and cytokine production in patients with multiple sclerosis. J. Intern. Med., 254: 363-374, 2003.
10. Lalive, P. H., Menge, T., Delarasse, C, Delia, G. B., Pham-Dinh, D., Villoslada, P., von Budingen, H. C, and Genain, C. P. Antibodies to native myelin oligodendrocyte glycoprotein are serologic markers of early inflammation in multiple sclerosis.
Proc.NatLAcad.Sci.U.S.A, 103: 2280-2285, 2006. 1 1. O'Connor, K. C, Chitnis, T., Griffin, D. E., Piyasirisilp, S., Bar-Or, A., Khoury, S., Wucherpfennig, K. W., and Hafler, D. A. Myelin basic protein-reactive autoantibodies in the serum and cerebrospinal fluid of multiple sclerosis patients are characterized by low-affinity interactions. J.Neuroimmunol., 136: 140-148, 2003.
12. Almeras, L., Lefranc, D., Drobecq, H., de Seze, J., Dubucquoi, S., Vermersch, P., and
Prin, L. New antigenic candidates in multiple sclerosis: identification by serological proteome analysis. Proteomics., 4: 2184-2194, 2004.
13. KoIIn, J., Ren, H. M., Da, R. R., Zhang, Y., Spillner, E., Olek, M., Hermanowicz, N., Hilgenberg, L. G., Smith, M. A., Van Den, N. S., and Qin, Y. Triosephosphate isomerase- and glyceraldehyde-3-phosphate dehydrogenase-reactive autoantibodies in the cerebrospinal fluid of patients with multiple sclerosis. J.Immunol., 177: 5652-5658, 2006.
14. Poser, C. M., Paty, D. W., Scheinberg, L., McDonald, W. I., Davis, F. A., Ebers, G. C, Johnson, K. P., Sibley, W. A., Silberberg, D. H., and Tourtellotte, W. W. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Ann. Neurol., 13: 227-
231 , 1983.
15. Somers, V., Govarts, C, Hellings, N., Hupperts, R., and Stinissen, P. Profiling the autoantibody repertoire by serological antigen selection. J.Autoimmun., 25: 223-228, 2005.
16. Marks, J. D., Hoogenboom, H. R., Bonnert, T. P., McCafferty, J., Griffiths, A. D., and
Winter, G. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J MoI Biol, 222: 581-97, 1991.
17. Somers, V. A., Brandwijk, R. J., Joosten, B., Moerkerk, P. T., Arends, J. W., Menheere, P., Pieterse, W. O., Claessen, A., Scheper, R. J., Hoogenboom, H. R., and Hufton, S. E. A panel of candidate tumor antigens in colorectal cancer revealed by the serological selection of a phage displayed cDNA expression library. J.Immunol., 169: 2772-2780, 2002.
18. Cortese, I., Tafi, R., Grimaldi, L. M., Martino, G., Nicosia, A., and Cortese, R. Identification of peptides specific for cerebrospinal fluid antibodies in multiple sclerosis by using phage libraries. Proc.Natl.Acad.Sci.U.S.A, 93: 11063-11067, 1996. 19. Archelos, J. J., Trotter, J., Previtali, S., Weissbrich, B., Toyka, K. V., and Hartung, H. P. Isolation and characterization of an oligodendrocyte precursor-derived B-cell epitope in multiple sclerosis. Ann. Neurol., 43: 15-24, 1998.
20. Owens, G. P., Burgoon, M. P., Devlin, M. E., and Gilden, D. H. Strategies to identify sequences or antigens unique to multiple sclerosis. Mult.Scler., 2: 184-194, 1996.
21. Cortese, I., Capone, S., Luchetti, S., Grimaldi, L. M., Nicosia, A., and Cortese, R. CSF- enriched antibodies do not share specificities among MS patients. Mult.Scler., 4: 118- 123, 1998.
22. Becker, K. G., Mattson, D. H., Powers, J. M., Gado, A. M., and Biddison, W. E. Analysis of a sequenced cDNA library from multiple sclerosis lesions. J.Neuroimmunol., 77: 27-
38, 1997.
23. Robinson, W. H., Fontoura, P., Lee, B. J., de Vegvar, H. E., Tom, J., Pedotti, R., DiGennaro, C. D., Mitchell, D. J., Fong, D., Ho, P. P., Ruiz, P. J., Maverakis, E., Stevens, D. B., Bernard, C. C, Martin, R., Kuchroo, V. K., van Noort, J. M., Genain, C. P., Amor, S., Olsson, T., Utz, P. J., Garren, H., and Steinman, L. Protein microarrays guide tolerizing DNA vaccine treatment of autoimmune encephalomyelitis. Nat.Biotechnol., 21: 1033-1039, 2003.
24. Neer, E. J., Schmidt, C. J., Nambudripad, R., and Smith, T. F. The ancient regulatory- protein family of WD-repeat proteins. Nature, 371: 297-300, 1994.
25. Smith, T. F., Gaitatzes, C, Saxena, K., and Neer, E. J. The WD repeat: a common architecture for diverse functions. Trends Biochem.Sci., 24: 181-185, 1999.
26. Smith, E. F. and Lefebvre, P. A. PF20 gene product contains WD repeats and localizes to the intermicrotubule bridges in Chlamydomonas flagella. MoI. Biol. Cell, 8: 455-467, 1997.
27. Strausberg, R. L., Feingold, E. A., Grouse, L. H., Derge, J. G., Klausner, R. D., Collins,
F. S., Wagner, L., Shenmen, C. M., Schuler, G. D., Altschul, S. F., Zeeberg, B., Buetow, K. H., Schaefer, C. F., Bhat, N. K., Hopkins, R. F., Jordan, H., Moore, T., Max, S. I., Wang, J., Hsieh, F., Diatchenko, L., Marusina, K., Farmer, A. A., Rubin, G. M., Hong, L., Stapleton, M., Soares, M. B., Bonaldo, M. F., Casavant, T. L., Scheetz, T. E., Brownstein, M. J., Usdin, T. B., Toshiyuki, S., Carninci, P., Prange, C, Raha, S. S.,
Loquellano, N. A., Peters, G. J., Abramson, R. D., Mullahy, S. J., Bosak, S. A., McEwan, P. J., McKernan, K. J., Malek, J. A., Gunaratne, P. H., Richards, S., Worley, K. C, Hale, S., Garcia, A. M., Gay, L. J., Hulyk, S. W., Villalon, D. K., Muzny, D. M., Sodergren, E. J., Lu, X., Gibbs, R. A., Fahey, J., Helton, E., Ketteman, M., Madan, A., Rodrigues, S., Sanchez, A., Whiting, M., Madan, A., Young, A. C, Shevchenko, Y., Bouffard, G. G., Blakesley, R. W., Touchman, J. W., Green, E. D., Dickson, M. C, Rodriguez, A. C, Grimwood, J., Schmutz, J., Myers, R. M., Butterfield, Y. S.,
Krzywinski, M. I., Skalska, U., Smailus, D. E., Schnerch, A., Schein, J. E., Jones, S. J., and Marra, M. A. Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. Proc.Natl.Acad.Sci.U.S.A, 99: 16899-16903, 2002.
28. Ota, T., Suzuki, Y., Nishikawa, T., Otsuki, T., Sugiyama, T., Irie, R., Wakamatsu, A., Hayashi, K., Sato, H., Nagai, K., Kimura, K., Makita, H., Sekine, M., Obayashi, M.,
Nishi, T., Shibahara, T., Tanaka, T., Ishii, S., Yamamoto, J., Saito, K., Kawai, Y., Isono, Y., Nakamura, Y., Nagahari, K., Murakami, K., Yasuda, T., Iwayanagi, T., Wagatsuma, M., Shiratori, A., Sudo, H., Hosoiri, T., Kaku, Y., Kodaira, H., Kondo, H., Sugawara, M., Takahashi, M., Kanda, K., Yokoi, T., Furuya, T., Kikkawa, E., Omura, Y., Abe, K., Kamihara, K., Katsuta, N., Sato, K., Tanikawa, M., Yamazaki, M., Ninomiya, K.,
Ishibashi, T., Yamashita, H., Murakawa, K., Fujimori, K., Tanai, H., Kimata, M., Watanabe, M., Hiraoka, S., Chiba, Y., Ishida, S., Ono, Y., Takiguchi, S., Watanabe, S., Yosida, M., Hotuta, T., Kusano, J., Kanehori, K., Takahashi-Fujii, A., Hara, H., Tanase, T. O., Nomura, Y., Togiya, S., Komai, F., Hara, R., Takeuchi, K., Arita, M., Imose, N., Musashino, K., Yuuki, H., Oshima, A., Sasaki, N., Aotsuka, S., Yoshikawa, Y.,
Matsunawa, H., lchihara, T., Shiohata, N., Sano, S., Moriya, S., Momiyama, H., Satoh, N., Takami, S., Terashima, Y., Suzuki, O., Nakagawa, S., Senoh, A., Mizoguchi, H., Goto, Y., Shimizu, F., Wakebe, H., Hishigaki, H., Watanabe, T., Sugiyama, A., Takemoto, M., Kawakami, B., Yamazaki, M., Watanabe, K., Kumagai, A., Itakura, S., Fukuzumi, Y., Fujimori, Y., Komiyama, M., Tashiro, H., Tanigami, A., Fujiwara, T., Ono,
T., Yamada, K., Fujii, Y., Ozaki, K., Hirao, M., Ohmori, Y., Kawabata, A., Hikiji, T., Kobatake, N., Inagaki, H., Ikema, Y., Okamoto, S., Okitani, R., Kawakami, T., Noguchi, S., Itoh, T., Shigeta, K., Senba, T., Matsumura, K., Nakajima, Y., Mizuno, T., Morinaga, M., Sasaki, M., Togashi, T., Oyama, M., Hata, H., Watanabe, M., Komatsu, T., Mizushima-Sugano, J., Satoh, T., Shirai, Y., Takahashi, Y., Nakagawa, K., Okumura,
K., Nagase, T., Nomura, N., Kikuchi, H., Masuho, Y., Yamashita, R., Nakai, K., Yada, T., Nakamura, Y., Ohara, O., Isogai, T., and Sugano, S. Complete sequencing and characterization of 21 ,243 full-length human cDNAs. Nat.Genet., 36: 40-45, 2004.
29. Ng, B., Yang, F., Huston, D. P., Yan, Y., Yang, Y., Xiong, Z., Peterson, L. E., Wang, H., and Yang, X. F. Increased noncanonical splicing of autoantigen transcripts provides the structural basis for expression of untolerized epitopes. J.AIIergy Clin. Immunol., 114: 1463-1470, 2004.
30. Yeo, G., Holste, D., Kreiman, G., and Burge, C. B. Variation in alternative splicing across human tissues. Genome Biol., 5: R74, 2004.
31. Larsen, J. E., Lund, O., and Nielsen, M. Improved method for predicting linear B-cell epitopes. Immunome.Res., 2: 2, 2006.
32. Cepok, S., Zhou, D., Srivastava, R., Nessler, S., Stei, S., Bussow, K., Sommer, N., and Hemmer, B. Identification of Epstein-Barr virus proteins as putative targets of the immune response in multiple sclerosis. J. Clin. Invest, 115: 1352-1360, 2005.
33. Wang, X., Yu, J., Sreekumar, A., Varambally, S., Shen, R., Giacherio, D., Mehra, R.,
Montie, J. E., Pienta, K. J., Sanda, M. G., Kantoff, P. W., Rubin, M. A., Wei, J. T., Ghosh, D., and Chinnaiyan, A. M. Autoantibody signatures in prostate cancer. N.Engl.J.Med., 353: 1224-1235, 2005.
34. Keegan, M., Konig, F., McClelland, R., Bruck, W., Morales, Y., Bitsch, A., Panitch, H., Lassmann, H., Weinshenker, B., Rodriguez, M., Parisi, J., and Lucchinetti, C. F.
Relation between humoral pathological changes in multiple sclerosis and response to therapeutic plasma exchange. Lancet, 366: 579-582, 2005.
35. Stuve, O., Cepok, S., Elias, B., Saleh, A., Hartung, H. P., Hemmer, B., and Kieseier, B. C. Clinical stabilization and effective B-lymphocyte depletion in the cerebrospinal fluid and peripheral blood of a patient with fulminant relapsing-remitting multiple sclerosis.
Arch. Neurol, 62: 1620-1623, 2005.

Claims

Claims
1. A composition comprising at least two different polypeptides comprising a sequence represented by any of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
2. The in vitro use of a composition according to claim 1 for detecting the presence and/or quantity of specific antibodies to at least one polypeptide of said composition in a body fluid of a mammal.
3. The use according to claim 2 wherein said mammal is a human.
4. The use according to claims 2 and 3 wherein said body fluid is cerebrospinal fluid.
5. The use according to claim 2-3 wherein said detection is via immune-enzymatic processes comprising enzyme-linked immunosorbant assays (ELISA), immunofluorescent techniques, radioimmunological assays (RIA), immunoblotting and
LINE blot.
6. The use according to claims 2-5 wherein the detection and/or quantity of at least one specific antibody is indicative for multiple sclerosis.
7. A method for specifically detecting multiple sclerosis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the presence or quantity of said antibody indicates that said mammal suffers from multiple sclerosis .
8. A method according to claim 7 wherein said mammal is a human.
9. A method according to claims 7 and 8 wherein said body fluid is cerebrospinal fluid.
10. A method according to claims 7-9 wherein said detection is via immune-enzymatic processes comprising enzyme-linked immunosorbant assays (ELISA), immunofluorescent techniques, radioimmunological assays (RIA) and immunoblotting and LINE blot.
1 1. A method according to claims 7-10 wherein the detection and/or quantity of at least one specific antibody is indicative for multiple sclerosis.
12. A method for evaluating the prognosis and/or disease severity of multiple sclerosis in a patient comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said patient wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO:
1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the decreased or increased concentration of said antibody indicates the prognosis of multiple sclerosis in said patient.
13. A method for selecting a patient for a specific therapeutic treatment of multiple sclerosis or evaluating the therapeutic treatment of multiple sclerosis in a patient comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said patient wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8 and wherein ii) the presence or quantity of said antibody leads to an election of a specific therapeutic treatment of multiple sclerosis in said patient.
14. A diagnostic kit for the detection of multiple sclerosis comprising a composition according to claim 1 , reagents for making a medium appropriate for an immunological reaction to occur and reagents enabling to detect the antigen/antibody complex which has been produced by said immunological reaction.
15. An antibody specifically binding to a polypeptide selected from the group comprising a polypeptide selected from the group consisting of SEQ ID NO: 1-8 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-8.
PCT/EP2008/054479 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis WO2008125651A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
CA002683865A CA2683865A1 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
MX2009011007A MX2009011007A (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis.
EP08749552A EP2145182A2 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
US12/595,597 US8772225B2 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
AU2008237878A AU2008237878A1 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
JP2010502532A JP2010523629A (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
CN200880018874A CN101680883A (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
ZA2009/07027A ZA200907027B (en) 2007-04-12 2009-10-08 Biomarkers for multiple sclerosis
NO20093327A NO20093327L (en) 2007-04-12 2009-11-11 Biomarkers for multiple sclerosis
US14/290,782 US20140273033A1 (en) 2007-04-12 2014-05-29 Biomarkers for multiple sclerosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP07106081.8 2007-04-12
EP07106081 2007-04-12

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US12/595,597 A-371-Of-International US8772225B2 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
EP12180809.1A Previously-Filed-Application EP2579038B1 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis
US14/290,782 Continuation-In-Part US20140273033A1 (en) 2007-04-12 2014-05-29 Biomarkers for multiple sclerosis

Publications (2)

Publication Number Publication Date
WO2008125651A2 true WO2008125651A2 (en) 2008-10-23
WO2008125651A3 WO2008125651A3 (en) 2009-03-12

Family

ID=39642898

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/054479 WO2008125651A2 (en) 2007-04-12 2008-04-14 Biomarkers for multiple sclerosis

Country Status (12)

Country Link
US (1) US8772225B2 (en)
EP (2) EP2145182A2 (en)
JP (1) JP2010523629A (en)
CN (1) CN101680883A (en)
AU (1) AU2008237878A1 (en)
CA (1) CA2683865A1 (en)
MX (1) MX2009011007A (en)
NO (1) NO20093327L (en)
PL (1) PL390236A1 (en)
RU (1) RU2009141523A (en)
WO (1) WO2008125651A2 (en)
ZA (1) ZA200907027B (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012012725A2 (en) 2010-07-23 2012-01-26 President And Fellows Of Harvard College Methods of detecting diseases or conditions using phagocytic cells
EP2437060A1 (en) * 2010-10-01 2012-04-04 Protagen AG Marker sequences for multiple sclerosis and use of same
JP2012508865A (en) * 2008-11-12 2012-04-12 イエダ リサーチ アンド ディベロプメント カンパニー リミテッド Diagnosis of multiple sclerosis
EP2569446A2 (en) * 2010-05-12 2013-03-20 Steven E. Schutzer Diagnostic markers for neuropsychiatric disease
WO2013135789A1 (en) * 2012-03-15 2013-09-19 Universiteit Hasselt Means and methods for the determination of the joint destruction progression rate in rheumatoid arthritis patients
WO2013188846A1 (en) 2012-06-15 2013-12-19 Harry Stylli Methods of detecting diseases or conditions
WO2013188828A1 (en) 2012-06-15 2013-12-19 Harry Stylli Methods of detecting diseases or conditions using circulating diseased cells
US10494675B2 (en) 2013-03-09 2019-12-03 Cell Mdx, Llc Methods of detecting cancer
US10626464B2 (en) 2014-09-11 2020-04-21 Cell Mdx, Llc Methods of detecting prostate cancer
WO2020121305A1 (en) 2018-12-12 2020-06-18 Hadasit Medical Research Services And Development Ltd. Markers of disease prognosis in multiple sclerosis
US10934589B2 (en) 2008-01-18 2021-03-02 President And Fellows Of Harvard College Methods of detecting signatures of disease or conditions in bodily fluids
US10961578B2 (en) 2010-07-23 2021-03-30 President And Fellows Of Harvard College Methods of detecting prenatal or pregnancy-related diseases or conditions
US11111537B2 (en) 2010-07-23 2021-09-07 President And Fellows Of Harvard College Methods of detecting autoimmune or immune-related diseases or conditions
US11585814B2 (en) 2013-03-09 2023-02-21 Immunis.Ai, Inc. Methods of detecting prostate cancer
EP4303584A2 (en) 2010-07-23 2024-01-10 President and Fellows of Harvard College Methods for detecting signatures of disease or conditions in bodily fluids

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140273033A1 (en) * 2007-04-12 2014-09-18 Universiteit Hasselt Biomarkers for multiple sclerosis
US11654988B2 (en) 2019-06-26 2023-05-23 Weel Autonomy Inc. Balancing system in an autonomous electronic bicycle
US10754340B1 (en) * 2019-06-26 2020-08-25 Weel Autonomy Inc. Virtual gearing in an autonomous electronic bicycle
CN117802214A (en) * 2022-09-30 2024-04-02 厦门英博迈生物科技有限公司 Marker for diagnosis of multiple sclerosis and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003000733A2 (en) 2001-06-22 2003-01-03 Universita' Degli Studi Di Firenze Glycopeptides, their preparation and use in the diagnosis or therapeutic treatment of multiple sclerosis
US20040043431A1 (en) 2002-08-29 2004-03-04 Aristo Vojdani Diagnosis of multiple sclerosis and other demyelinating diseases
WO2004034031A2 (en) 2002-10-11 2004-04-22 The Regents Of The University Of California A method for diagnosis and prognosis of multiple sclerosis

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030092089A1 (en) * 2001-11-14 2003-05-15 Moscarello Mario Anthony Method for diagnosing multiple sclerosis and an assay therefore

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003000733A2 (en) 2001-06-22 2003-01-03 Universita' Degli Studi Di Firenze Glycopeptides, their preparation and use in the diagnosis or therapeutic treatment of multiple sclerosis
US20040043431A1 (en) 2002-08-29 2004-03-04 Aristo Vojdani Diagnosis of multiple sclerosis and other demyelinating diseases
WO2004034031A2 (en) 2002-10-11 2004-04-22 The Regents Of The University Of California A method for diagnosis and prognosis of multiple sclerosis

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
ALMERAS, L.; LEFRANC, D.; DROBECQ, H.; DE SEZE, J.; DUBUCQUOI, S.; VERMERSCH, P.; PRIN, L.: "New antigenic candidates in multiple sclerosis: identification by serological proteome analysis", PROTEOMICS, vol. 4, 2004, pages 2184 - 2194
ARCHELOS, J. J.; TROTTER, J.; PREVITALI, S.; WEISSBRICH, B.; TOYKA, K. V.; HARTUNG, H. P.: "Isolation and characterization of an oligodendrocyte precursor-derived B-cell epitope in multiple sclerosis", ANN.NEUROL., vol. 43, 1998, pages 15 - 24
ATHERTON; SHEPARD: "Solid phase peptide synthesis", 1989, IRL PRESS
ATHERTON; SHEPPARD: "Solid Phase Peptide Synthesis", 1989, IRL PRESS
BARANZINI, S. E.; JEONG, M. C.; BUTUNOI, C.; MURRAY, R. S.; BERNARD, C. C.; OKSENBERG, J. R.: "B cell repertoire diversity and clonal expansion in multiple sclerosis brain lesions", J.IMMUNOL., vol. 163, 1999, pages 5133 - 5144
BECKER, K. G.; MATTSON, D. H.; POWERS, J. M.; GADO, A. M.; BIDDISON, W. E.: "Analysis of a sequenced cDNA library from multiple sclerosis lesions", J.NEUROIMMUNOL., vol. 77, 1997, pages 27 - 38
CEPOK, S.; ZHOU, D.; SRIVASTAVA, R.; NESSLER, S.; STEI, S.; BUSSOW, K.; SOMMER, N.; HEMMER, B.: "Identification of Epstein-Barr virus proteins as putative targets of the immune response in multiple sclerosis", J.CLIN.LNVEST, vol. 115, 2005, pages 1352 - 1360
CORTESE, I.; CAPONE, S.; LUCHETTI, S.; GRIMALDI, L. M.; NICOSIA, A.; CORTESE, R: "CSF- enriched antibodies do not share specificities among MS patients", MULT.SCLER., vol. 4, 1998, pages 118 - 123
CORTESE, I.; TAFI, R.; GRIMALDI, L. M.; MARTINO, G.; NICOSIA, A.; CORTESE, R.: "Identification of peptides specific for cerebrospinal fluid antibodies in multiple sclerosis by using phage libraries", PROC.NATI.ACAD.SCI.U.S.A, vol. 93, 1996, pages 11063 - 11067
CROSS, A. H.; TROTTER, J. L.; LYONS, J.: "B cells and antibodies in CNS demyelinating disease", J.NEUROIMMUNOL., vol. 112, 2001, pages 1 - 14
GENAIN, C. P.; CANNELLA, B.; HAUSER, S. L.; RAINE, C. S.: "Identification of autoantibodies associated with myelin damage in multiple sclerosis", NAT.MED., vol. 5, 1999, pages 170 - 175
HOUBENWEYL: "Methode der organischen chemie", vol. 15-1, II, 1974, THIEME
KARNI, A.; BAKIMER-KLEINER, R.; ABRAMSKY, O.; BEN NUN, A.: "Elevated levels of antibody to myelin oligodendrocyte glycoprotein is not specific for patients with multiple sclerosis", ARCH.NEUROL., vol. 56, 1999, pages 311 - 315
KEEGAN, M.; KONIG, F.; MCCLELLAND, R.; BRUCK, W.; MORALES, Y.; BITSCH, A.; PANITCH, H.; LASSMANN, H.; WEINSHENKER, B.; RODRIGUEZ,: "Relation between humoral pathological changes in multiple sclerosis and response to therapeutic plasma exchange", LANCET, vol. 366, 2005, pages 579 - 582
KOHLER, G; MILSTEIN, C, NATURE, vol. 256, 1973, pages 495 - 497
KOLLN, J.; REN, H. M.; DA, R. R.; ZHANG, Y.; SPILLNER, E.; OLEK, M.; HERMANOWICZ, N.; HILGENBERG, L. G.; SMITH, M. A.; VAN DEN, N.: "Triosephosphate isomerase- and glyceraldehyde-3-phosphate dehydrogenase-reactive autoantibodies in the cerebrospinal fluid of patients with multiple sclerosis", J.IMMUNOL., vol. 177, 2006, pages 5652 - 5658
LALIVE, P. H.; MENGE, T.; DELARASSE, C.; DELLA, G. B.; PHAM-DINH, D.; VILLOSLADA, P.; VON BUDINGEN, H. C.; GENAIN, C. P.: "Antibodies to native myelin oligodendrocyte glycoprotein are serologic markers of early inflammation in multiple sclerosis", PROC.NATI.ACAD.SCI.U.S.A, vol. 103, 2006, pages 2280 - 2285
LARSEN, J. E.; LUND, O.; NIELSEN, M.: "Improved method for predicting linear B-cell epitopes", IMMUNOME.RES., vol. 2, 2006, pages 2
LUCCHINETTI, C. F.; BRUCK, W.; RODRIGUEZ, M.; LASSMANN, H.: "Distinct patterns of multiple sclerosis pathology indicates heterogeneity on pathogenesis", BRAIN PATHOL., vol. 6, 1996, pages 259 - 274
LUCCHINETTI, C.; BRUCK, W.; PARISI, J.; SCHEITHAUER, B.; RODRIGUEZ, M.; LASSMANN, H.: "Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination", ANN.NEUROL., vol. 47, 2000, pages 707 - 717
MARKS, J. D.; HOOGENBOOM, H. R.; BONNERT, T. P.; MCCAFFERTY, J.; GRIFFITHS, A. D.; WINTER, G.: "By-passing immunization. Human antibodies from V-gene libraries displayed on phage", J MOL BIOL, vol. 222, 1991, pages 581 - 97
MOLLER, J. R.; JOHNSON, D.; BRADY, R. O.; TOURTELLOTTE, W. W.; QUARLES, R. H.: "Antibodies to myelin-associated glycoprotein (MAG) in the cerebrospinal fluid of multiple sclerosis patients", J.NEUROIMMUNOL., vol. 22, 1989, pages 55 - 61
NEER, E. J.; SCHMIDT, C. J.; NAMBUDRIPAD, R.; SMITH, T. F.: "The ancient regulatory- protein family of WD-repeat proteins", NATURE, vol. 371, 1994, pages 297 - 300
NG, B.; YANG, F.; HUSTON, D. P.; YAN, Y.; YANG, Y.; XIONG, Z.; PETERSON, L. E.; WANG, H.; YANG, X. F.: "Increased noncanonical splicing of autoantigen transcripts provides the structural basis for expression of untolerized epitopes", J.ALLERGY CLIN.LMMUNOL., vol. 114, 2004, pages 1463 - 1470
O'CONNOR, K. C.; CHITNIS, T.; GRIFFIN, D. E.; PIYASIRISILP, S.; BAR-OR, A.; KHOURY, S.; WUCHERPFENNIG, K. W.; HAFLER, D. A.: "Myelin basic protein-reactive autoantibodies in the serum and cerebrospinal fluid of multiple sclerosis patients are characterized by low-affinity interactions", J.NEUROIMMUNOL., vol. 136, 2003, pages 140 - 148
OTA, T.; SUZUKI, Y.; NISHIKAWA, T.; OTSUKI, T.; SUGIYAMA, T.; IRIE, R.; WAKAMATSU, A.; HAYASHI, K.; SATO, H.; NAGAI, K.: "Complete sequencing and characterization of 21,243 full-length human cDNAs", NAT.GENET., vol. 36, 2004, pages 40 - 45
OWENS, G. P.; BURGOON, M. P.; DEVLIN, M. E.; GILDEN, D. H.: "Strategies to identify sequences or antigens unique to multiple sclerosis", MULT.SCLER., vol. 2, 1996, pages 184 - 194
POSER, C. M.; PATY, D. W.; SCHEINBERG, L.; MCDONALD, W. I.; DAVIS, F. A.; EBERS, G. C.; JOHNSON, K. P.; SIBLEY, W. A.; SILBERBERG,: "New diagnostic criteria for multiple sclerosis: guidelines for research protocols", ANN.NEUROL., vol. 13, 1983, pages 227 - 231
QIN, Y.; DUQUETTE, P.; ZHANG, Y.; TALBOT, P.; POOLE, R.; ANTEL, J.: "Clonal expansion and somatic hypermutation of V(H) genes of B cells from cerebrospinal fluid in multiple sclerosis", J.CLIN.LNVEST, vol. 102, 1998, pages 1045 - 1050
ROBINSON, W. H.; FONTOURA, P.; LEE, B. J.; DE VEGVAR, H. E.; TOM, J.; PEDOTTI, R.; DIGENNARO, C. D.; MITCHELL, D. J.; FONG, D.; HO: "Protein microarrays guide tolerizing DNA vaccine treatment of autoimmune encephalomyelitis", NAT.BIOTECHNOL., vol. 21, 2003, pages 1033 - 1039
SMITH, E. F.; LEFEBVRE, P. A.: "PF20 gene product contains WD repeats and localizes to the intermicrotubule bridges in Chlamydomonas flagella", MOL.BIOL.CELL, vol. 8, 1997, pages 455 - 467
SMITH, T. F.; GAITATZES, C.; SAXENA, K.; NEER, E. J.: "The WD repeat: a common architecture for diverse functions", TRENDS BIOCHEM.SCI., vol. 24, 1999, pages 181 - 185
SOMERS V. ET AL., J. OF AUTOIMMUNITY, vol. 25, 2005, pages 223 - 228
SOMERS, V. A.; BRANDWIJK, R. J.; JOOSTEN, B.; MOERKERK, P. T.; ARENDS, J. W.; MENHEERE, P.; PIETERSE, W. O.; CLAESSEN, A.; SCHEPER: "A panel of candidate tumor antigens in colorectal cancer revealed by the serological selection of a phage displayed cDNA expression library", J.IMMUNOL., vol. 169, 2002, pages 2772 - 2780
SOMERS, V.; GOVARTS, C.; HELLINGS, N.; HUPPERTS, R.; STINISSEN, P.: "Profiling the autoantibody repertoire by serological antigen selection", J.AUTOIMMUN., vol. 25, pages 223 - 228
STEWART; YOUNG: "Solid Phase Peptide Synthesis", 1984, PIERCE CHEMICAL COMPANY
STRAUSBERG, R. L.; FEINGOLD, E. A.; GROUSE, L. H.; DERGE, J. G.; KLAUSNER, R. D.; COLLINS, F. S.; WAGNER, L.; SHENMEN, C. M.; SCHU: "Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences", PROC.NATI.ACAD.SCI.U.S.A, vol. 99, 2002, pages 16899 - 16903
STUVE, O.; CEPOK, S.; ELIAS, B.; SALEH, A.; HARTUNG, H. P.; HEMMER, B.; KIESEIER, B. C.: "Clinical stabilization and effective B-ymphocyte depletion in the cerebrospinal fluid and peripheral blood of a patient with fulminant relapsing-remitting multiple sclerosis", ARCH.NEUROL, vol. 62, 2005, pages 1620 - 1623
TOWBIN H. ET AL.: "Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications", PROC. NATL. ACAD. SCI. USA, vol. 76, 1979, pages 4350 - 4354
VOJDANI, A.; VOJDANI, E.; COOPER, E.: "Antibodies to myelin basic protein, myelin oligodendrocytes peptides, alpha-beta-crystallin, lymphocyte activation and cytokine production in patients with multiple sclerosis", J.INTERN.MED., vol. 254, 2003, pages 363 - 374
WANG, X.; YU, J.; SREEKUMAR, A.; VARAMBALLY, S.; SHEN, R.; GIACHERIO, D.; MEHRA, R.; MONTIE, J. E.; PIENTA, K. J.; SANDA, M. G.: "Autoantibody signatures in prostate cancer", N.ENGL.J.MED., vol. 353, 2005, pages 1224 - 1235
YEO, G.; HOLSTE, D.; KREIMAN, G.; BURGE, C. B.: "Variation in alternative splicing across human tissues", GENOME BIOL., vol. 5, 2004, pages R74

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10934589B2 (en) 2008-01-18 2021-03-02 President And Fellows Of Harvard College Methods of detecting signatures of disease or conditions in bodily fluids
US11001894B2 (en) 2008-01-18 2021-05-11 President And Fellows Of Harvard College Methods of detecting signatures of disease or conditions in bodily fluids
US10934588B2 (en) 2008-01-18 2021-03-02 President And Fellows Of Harvard College Methods of detecting signatures of disease or conditions in bodily fluids
JP2012508865A (en) * 2008-11-12 2012-04-12 イエダ リサーチ アンド ディベロプメント カンパニー リミテッド Diagnosis of multiple sclerosis
US9267945B2 (en) 2008-11-12 2016-02-23 Yeda Research And Development Co. Ltd. Diagnosis of multiple sclerosis
EP2569446A4 (en) * 2010-05-12 2014-05-21 Steven E Schutzer Diagnostic markers for neuropsychiatric disease
AU2019204118B2 (en) * 2010-05-12 2021-04-22 Steven E. Schutzer Diagnostic markers for neuropsychiatric disease
EP2569446A2 (en) * 2010-05-12 2013-03-20 Steven E. Schutzer Diagnostic markers for neuropsychiatric disease
EP4303584A2 (en) 2010-07-23 2024-01-10 President and Fellows of Harvard College Methods for detecting signatures of disease or conditions in bodily fluids
US11111537B2 (en) 2010-07-23 2021-09-07 President And Fellows Of Harvard College Methods of detecting autoimmune or immune-related diseases or conditions
WO2012012725A2 (en) 2010-07-23 2012-01-26 President And Fellows Of Harvard College Methods of detecting diseases or conditions using phagocytic cells
US10961578B2 (en) 2010-07-23 2021-03-30 President And Fellows Of Harvard College Methods of detecting prenatal or pregnancy-related diseases or conditions
WO2012042062A3 (en) * 2010-10-01 2012-06-21 Protagen Ag Marker sequences for multiple sclerosis and use thereof
EP2437060A1 (en) * 2010-10-01 2012-04-04 Protagen AG Marker sequences for multiple sclerosis and use of same
WO2013135789A1 (en) * 2012-03-15 2013-09-19 Universiteit Hasselt Means and methods for the determination of the joint destruction progression rate in rheumatoid arthritis patients
WO2013188828A1 (en) 2012-06-15 2013-12-19 Harry Stylli Methods of detecting diseases or conditions using circulating diseased cells
WO2013188846A1 (en) 2012-06-15 2013-12-19 Harry Stylli Methods of detecting diseases or conditions
US10494675B2 (en) 2013-03-09 2019-12-03 Cell Mdx, Llc Methods of detecting cancer
US11585814B2 (en) 2013-03-09 2023-02-21 Immunis.Ai, Inc. Methods of detecting prostate cancer
US10626464B2 (en) 2014-09-11 2020-04-21 Cell Mdx, Llc Methods of detecting prostate cancer
WO2020121305A1 (en) 2018-12-12 2020-06-18 Hadasit Medical Research Services And Development Ltd. Markers of disease prognosis in multiple sclerosis

Also Published As

Publication number Publication date
WO2008125651A3 (en) 2009-03-12
EP2579038A2 (en) 2013-04-10
EP2579038A3 (en) 2013-07-03
EP2579038B1 (en) 2014-10-01
US8772225B2 (en) 2014-07-08
JP2010523629A (en) 2010-07-15
CN101680883A (en) 2010-03-24
AU2008237878A1 (en) 2008-10-23
PL390236A1 (en) 2010-07-05
EP2145182A2 (en) 2010-01-20
MX2009011007A (en) 2009-12-04
CA2683865A1 (en) 2008-10-23
RU2009141523A (en) 2011-05-20
ZA200907027B (en) 2010-12-29
US20100297678A1 (en) 2010-11-25
NO20093327L (en) 2009-12-02

Similar Documents

Publication Publication Date Title
US8772225B2 (en) Biomarkers for multiple sclerosis
US11199549B2 (en) MEl&#39;hods and means for diagnosing spondylarthritis using autoantibody markers
Hadjivassiliou et al. Autoantibodies in gluten ataxia recognize a novel neuronal transglutaminase
Somers et al. Autoantibody profiling in multiple sclerosis reveals novel antigenic candidates
US20180030122A1 (en) Biomarkers for rheumatoid arthritis
US20120046181A1 (en) Biomarkers for the Diagnosis of Renal Allograft and Kidney Status
US11609237B2 (en) Marker sequences for diagnosing and stratifying systemic sclerosis patients
JP2015528912A (en) How to diagnose scleroderma
JP2014516155A (en) Methods for diagnosis of early rheumatoid arthritis
US20140273033A1 (en) Biomarkers for multiple sclerosis
JP6663000B2 (en) Autoantigens for the diagnosis of rheumatoid arthritis
US20220365082A1 (en) Method and means for diagnosis of spondyloarthritis
US9915667B2 (en) Methods and means for diagnosing vasculitis
AU2015280271B2 (en) Compositions and methods for the diagnosis of systemic autoimmune disease
EP2816354B1 (en) Biomarker for complex regional pain syndrome (CRPS)
JP2006071409A (en) Sjogren&#39;s syndrome diagnostic kit
WO2020035123A1 (en) Autoantibodies binding to negative elongation factor e (nelf-e) for diagnosing sarcoidosis

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880018874.2

Country of ref document: CN

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

Ref document number: 08749552

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2010502532

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2683865

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 390236

Country of ref document: PL

Ref document number: MX/A/2009/011007

Country of ref document: MX

Ref document number: 12009501935

Country of ref document: PH

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 580377

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2008237878

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 6752/DELNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008749552

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009141523

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 12595597

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: PI0810173

Country of ref document: BR

Free format text: REGULARIZE A PROCURACAO, UMA VEZ QUE A PROCURACAO APRESENTADA NAO POSSUI DATA.

ENPW Started to enter national phase and was withdrawn or failed for other reasons

Ref document number: PI0810173

Country of ref document: BR

Free format text: PEDIDO RETIRADO EM RELACAO AO BRASIL POR NAO ATENDER AS DETERMINACOES REFERENTES A ENTRADA DO PEDIDO NA FASE NACIONAL E POR NAO CUMPRIMENTO DA EXIGENCIA FORMULADA NA RPI NO 2287 DE 04/11/2014.