EP1354199A2 - Diagnostic et traitement de la sclerose en plaques - Google Patents

Diagnostic et traitement de la sclerose en plaques

Info

Publication number
EP1354199A2
EP1354199A2 EP02715572A EP02715572A EP1354199A2 EP 1354199 A2 EP1354199 A2 EP 1354199A2 EP 02715572 A EP02715572 A EP 02715572A EP 02715572 A EP02715572 A EP 02715572A EP 1354199 A2 EP1354199 A2 EP 1354199A2
Authority
EP
European Patent Office
Prior art keywords
mspi
msf
multiple sclerosis
fragment
mspis
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP02715572A
Other languages
German (de)
English (en)
Inventor
Herath Mudiyanselage Athula Chandrasiri Herath
Rajesh Bhikhu Parekh
Christian Rohlff
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oxford Glycosciences UK Ltd
Original Assignee
Oxford Glycosciences UK Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxford Glycosciences UK Ltd filed Critical Oxford Glycosciences UK Ltd
Publication of EP1354199A2 publication Critical patent/EP1354199A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4713Autoimmune diseases, e.g. Insulin-dependent diabetes mellitus, multiple sclerosis, rheumathoid arthritis, systemic lupus erythematosus; Autoantigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • PROTEINS GENES AND THEIR USE FOR DIAGNOSIS AND TREATMENT OF MULTIPLE SCLEROSIS
  • the present invention relates to the identification of polypeptides, proteins and protein isoforms that are associated with Multiple Sclerosis and its onset and development, and of genes encoding the same, and to their use for e.g., clinical screening, diagnosis, prognosis, therapy and prophylaxis, as well as for drug screening and drug development.
  • MS Multiple sclerosis
  • CNS central nervous system
  • Proteomic approaches appear most suitable for a molecular dissection of such disease phenotypes in the CNS.
  • the entire CNS is largely inaccessible to meaningful mRNA expression-based analyses of primary human material, since post mortem delays in primary human brain tissue affects mRNAs more readily than proteins (Edgar et al. Molecular Psychiatry 1999, 4, 173-17).
  • CSF cerebrospinal fluid
  • changes in its protein composition may reveal alterations in CNS protein expression pattern causatively or diagnostically linked to the disease.
  • Reasonable amounts of disease specific proteins (DSPs) are secreted or released into body fluids by diseased tissue in the living patient at the onset and/or during progression of the disease.
  • MS MS
  • Poser criteria include abnormalities of evoked response and Magnetic Resonance Imaging (MRI), and immunologic abnormalities in the CSF (Poser, CM. et al. (1983) Ann Neurol 13: 227-231).
  • Symptoms of MS at presentation vary among studied populations but include sensory symptoms in 24% of patients, optic neuritis in of 31% of patients, limb weakness in 17% of patients and brain stem and cerebellar symptoms 25% of patients. (Thompson, A.J. et al. (1986) Q.J. Med. 225:69-80).
  • MS has a wide range of clinical presentations and courses, and the clinical course of any given patient is unpredictable.
  • MS patients it begins with a relapsing and remitting course, where episodes of neurological dysfunction last several weeks. Over the course of the disease remissions tend be less than complete and patients pass into a progressive phase (secondary progression).
  • secondary progression During this phase of the disease patients develop severe irreversible disabilities. About one-third of patients have benign MS, which does not develop secondary progression. Approximately 10% of patients develop progressive disability from onset without relapses and remissions (primary progressive MS). Few biochemical changes have been identified in MS. Consequently the identification and characterization of cellular and/or molecular causative defects and neuropathologies are necessary for improved treatment of neurological disorders. Due to the possibility of worsening or recurrence, speedy diagnosis would be of great benefit, in particular to categorise the patient as follows: 1. Benign versus progressive MS 2. Primary versus secondary progressive MS
  • MS has no objective biochemical markers useful for diagnosis and prognosis in living patients.
  • the identification of DSPs in the CSF of MS patients may provide important insights into disease pathology and opportunities for better diagnosis and treatment strategies.
  • Isoelectric focusing of CSF from MS patients revealed the presence of oligoclonal bands in 95% of patients with MS (McLean et al. (1990) Brain, 113:1269-89).
  • this finding is not specific to MS patients and can also be detected in other neurological disorders including Guillain-Barre syndrome, sarcoidosis and chronic menengitis. Therefore, the specificity and the sensitivity of distinguishing individual neurological disorders as well as acute and chronic CNS disease may require the selection of a repertoire of DSPs rather than an individual protein.
  • the present invention provides methods and compositions for clinical screening, diagnosis and treatment of MS, for monitoring the effectiveness of Multiple Sclerosis treatment, for selecting participants in clinical trials, for identifying patients most likely to respond to a particular therapeutic treatment and for screening and development of drugs for treatment of MS.
  • a first aspect of the invention provides methods for diagnosis of MS that comprise analyzing a sample of CSF by two-dimensional electrophoresis to detect the presence or level of at least one Multiple Sclerosis-Associated Feature (MSF), e.g., one or more of the MSFs disclosed herein or any combination thereof.
  • MSF Multiple Sclerosis-Associated Feature
  • a second aspect of the invention provides methods for diagnosis of MS that comprise detecting in a sample of CSF the presence or level of at least one Multiple Sclerosis-Associated Protein Isoform (MSPI), e.g., one or more of the MSPIs disclosed herein or any combination thereof.
  • MSPI Multiple Sclerosis-Associated Protein Isoform
  • a third aspect of the invention provides antibodies, e.g. monoclonal and polyclonal, chimeric and humanised antibodies capable of immunospecific binding to an MSPI, e.g., an MSPI disclosed herein.
  • a fourth aspect of the invention provides a preparation comprising an isolated MSPI, i.e., an MSPI free from polypeptides, proteins or protein isoforms having a significantly different isoelectric point or a significantly different apparent molecular weight from the MSPI.
  • a fifth aspect of the invention provides kits that may be used in the above recited methods and that may comprise single or multiple preparations, or antibodies, together with other reagents, labels, substrates, if needed, and directions for use. The kits may be used for diagnosis of disease, or may be assays for the identification of new diagnostic and/or therapeutic agents.
  • a sixth aspect of the invention provides methods of treating MS, comprising administering to a subject a therapeutically effective amount of an agent that modulates (e.g., upregulates or downregulates) the expression or activity (e.g. enzymatic or binding activity), or both, of an MSPI in subjects having MS, in order to prevent or delay the onset or development of MS, to prevent or delay the progression of MS, or to ameliorate the symptoms of MS.
  • a seventh aspect of the invention provides methods of screening for agents that modulate (e.g., upregulate or downregulate) a characteristic of, e.g., the expression or the enzymatic or binding activity, of a MSF, MSPI, a MSPI analog, or a MSPI-related polypeptide.
  • Figure 1 is an image obtained from 2-dimensional electrophoresis of CSF, which has been annotated to identify twelve landmark features, designated CSF1 to CSF12.
  • FIG. 2 is a flow chart depicting the characterization of an MSF and relationship of a MSF and MSPI.
  • a MSF may be further characterized as or by an MSPI having a particular peptide sequence associated with its pi and MW.
  • a MSF may comprise one or more MSPIs, which have indistinguishable pis and MWs using the Preferred Technology, but which have distinct peptide sequences.
  • the peptide sequence(s) of the MSPI can be utilized to search database(s) for previously identified proteins comprising such peptide sequence(s). It can be ascertained whether a commercially available antibody exists that may recognize the previously identified protein and/or a member of its protein family.
  • the invention described in detail below provides methods and compositions for clinical screening and diagnosis of Multiple Sclerosis in a mammalian subject for identifying patients most likely to respond to a particular therapeutic treatment, for monitoring the results of Multiple Sclerosis therapy, for drug screening and drug development.
  • the invention also encompasses the administration of therapeutic compositions to a mammalian subject to treat or prevent Multiple Sclerosis.
  • the mammalian subject may be a non-human mammal, but is preferably human, more preferably a human adult, i.e. a human subject at least 21 years old.
  • the invention will be described with respect to the analysis of CSF samples.
  • the assays and techniques described below can be applied to other types of samples, including a body fluid (for example but without limitation: blood, serum, plasma, saliva or urine), a tissue sample from a subject at risk of having or developing MS (e.g. a biopsy such as a brain biopsy) or homogenate thereof.
  • a body fluid for example but without limitation: blood, serum, plasma, saliva or urine
  • tissue sample from a subject at risk of having or developing MS e.g. a biopsy such as a brain biopsy
  • homogenate thereof e.g. a biopsy such as a brain biopsy
  • Feature refers to a spot identified in a 2D gel
  • MSF Multiple Sclerosis - Associated Feature
  • a feature or spot identified in a 2D gel is characterized by its isoelectric point (pi) and apparent molecular weight (MW) as determined by 2D gel electrophoresis, particularly utilizing the Preferred Technology described herein.
  • a feature is "differentially present" in a first sample or sample set with respect to a second sample or sample set when a method for detecting the said feature (e.g., 2D electrophoresis) gives a different signal when applied to the first and second samples or sample sets.
  • a MSF (or a Protein Isoform, i.e. MSPI, as defined infra) is "increased" in the first sample or sample set with respect to the second sample or sample set if the method of detection indicates that the MSF, or MSPI is more abundant in the first sample or sample set than in the second sample or sample set, or if the MSF, or MSPI is detectable in the first sample or sample set and substantially undetectable in the second sample or sample set.
  • a MSF, or MSPI is "decreased" in the first sample or sample set with respect to the second sample or sample set if the method of detection indicates that the MSF, or MSPI is less abundant in the first sample or sample set than in the second sample or sample set or if the MSF, or MSPI is undetectable in the first sample or sample set and detectable in the second sample or sample set.
  • the relative abundance of a feature in the two samples or sample sets is determined in reference to its normalized signal, in two steps.
  • the signal obtained upon detecting the feature in a first sample or sample set is normalized by reference to a suitable background parameter, e.g., (a) to the total protein in the sample being analyzed (e.g., total protein loaded onto a gel); (b) to an Expression Reference Feature (ERF) i.e., a feature whose abundance is substantially invariant, within the limits of variability of the Preferred Technology, in the population of subjects being examined, e.g. the ERFs disclosed in Table in, or (c) more preferably to the total signal detected as the sum of each of all proteins in the sample.
  • a suitable background parameter e.g., (a) to the total protein in the sample being analyzed (e.g., total protein loaded onto a gel); (b) to an Expression Reference Feature (ERF) i.e., a feature whose abundance is substantially invariant, within the limits of
  • the normalized signal for the feature in the first sample or sample set is compared with the normalized signal for the same feature in the second sample or sample set in order to identify features that are "differentially present" in the first sample or sample set with respect to the second sample or sample set.
  • “Fold change” includes “fold increase” and “fold decrease” and refers to the relative increase or decrease in abundance of a MSF or the relative increase or decrease in expression or activity of a polypeptide (e.g. a MSPI, as defined infra.) in a first sample or sample set compared to a second sample or sample set.
  • a MSF or polypeptide fold change may be measured by any technique known to those of skill in the art, albeit the observed increase or decrease will vary depending upon the technique used.
  • fold change is determined herein as described in the Examples infra.
  • MSPI Multiple Sclerosis-Associated Protein Isoform
  • a MSPI refers to a polypeptide that is differentially present in a first sample or sample set from a subject having MS compared with a second sample or sample set from a subject free from MS.
  • a MSPI is “differentially present” in a first sample or sample set with respect to a second sample or sample set when a method for detecting the said feature, (e.g. , 2D electrophoresis or immunoassay) gives a different signal when applied to the first and second samples or sample sets (as described above in relation to MSFs).
  • MSPI is characterised by one or more peptide sequences of which it is comprised, and further by a pi and MW, preferably determined by 2D electrophoresis, particularly utilising the Preferred Technology as described herein.
  • MSPIs are identified or characterised by the amino acid sequencing of MSFs ( Figure 2).
  • An MSPI is characterized as, or by, a particular peptide sequence associated with its pi and MW.
  • a MSF may comprise one or more MSPI(s), which have indistinguishable pis and MWs using the Preferred Technology, but which have distinct peptide sequences.
  • the peptide sequence(s) of the MSPI can be utilized to search database(s) for previously identified proteins comprising such peptide sequence(s). In some instances, it can be ascertained whether a commercially available antibody exists which may recognize the previously identified protein and/or a variant thereof.
  • the MSPI corresponds to the previously identified protein, or be a variant of the previously identified protein.
  • Variant refers to a polypeptide which is a member of a family of polypeptides that are encoded by a single gene or from a gene sequence within a family of related genes and which differ in their pi or MW, or both. Such variants can differ in their amino acid composition (e.g. as a result of alternative mRNA or premRNA processing, e.g. alternative splicing or limited proteolysis) and in addition, or in the alternative, may arise from differential post-translational modification (e.g., glycosylation, acylation, phosphorylation).
  • differential post-translational modification e.g., glycosylation, acylation, phosphorylation
  • Modulate in reference to expression or activity of a MSF, MSPI or MSPI-related polypeptide refers to any change, e.g., upregulation or downregulation, increase or decrease, of the expression or activity of the MSF, MSPI or MSPI-related polypeptide.
  • upregulation or downregulation increase or decrease, of the expression or activity of the MSF, MSPI or MSPI-related polypeptide.
  • modulation can be determined by assays known to those of skill in the art.
  • MSPI analog refers to a polypeptide that possesses similar or identical function(s) as a MSPI but need not necessarily comprise an amino acid sequence that is similar or identical to the amino acid sequence of the MSPI, or possess a structure that is similar or identical to that of the MSPI.
  • an amino acid sequence of a polypeptide is "similar" to that of a MSPI if it satisfies at least one of the following criteria: (a) the polypeptide has an amino acid sequence that is at least 30% (more preferably, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 99%) identical to the amino acid sequence of the MSPI; (b) the polypeptide is encoded by a nucleotide sequence that hybridizes under stringent conditions to a nucleotide sequence encoding at least 5 amino acid residues (more preferably, at least 10 amino acid residues, at least 15 amino acid residues, at least 20 amino acid residues, at least 25 amino acid residues, at least 40 amino acid residues, at least 50 amino acid residues, at least 60 amino residues, at least 70 amino acid residues, at
  • a polypeptide with "similar structure" to that of a MSPI refers to a polypeptide that has a similar secondary, tertiary or quarternary structure as that of the MSPI.
  • the structure of a polypeptide can determined by methods known to those skilled in the art, including but not limited to, X-ray crystallography, nuclear magnetic resonance, and crystallographic electron microscopy.
  • MSPI fusion protein refers to a polypeptide that comprises (i) an amino acid sequence of an MSPI, MSPI fragment, MSPI-related polypeptide or a fragment of an
  • MSPI-related polypeptide and (ii) an amino acid sequence of a heterologous polypeptide (i.e., a non-MSPI, non-MSPI fragment or non-MSPI-related polypeptide).
  • a heterologous polypeptide i.e., a non-MSPI, non-MSPI fragment or non-MSPI-related polypeptide.
  • MSPI homolog refers to a polypeptide that comprises an amino acid sequence similar to that of a MSPI but does not necessarily possess a similar or identical function as the MSPI.
  • MSPI ortholog refers to a non-human polypeptide that (i) comprises an amino acid sequence similar to that of a MSPI and (ii) possesses a similar or identical function to that of the MSPI.
  • MSPI-related polypeptide refers to a MSPI homolog, a MSPI analog, a variant of
  • MSPI MSPI, a MSPI ortholog, or any combination thereof.
  • Chimeric Antibody refers to a molecule in which different portions are derived from different animal species, such as those having a human immunoglobulin constant region and a variable region derived from a murine mAb. (See, e.g., Cabilly et al., U.S. Patent No. 4,816,567; and Boss et al., U.S. Patent No. 4,816397, which are incorporated herein by reference in their entirety.) ).
  • a portion of the antibody may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof) resulting in chimeric antibodies.
  • Humanised Antibody refers to a molecule from non-human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule.
  • CDRs complementarily determining regions
  • “Derivative” refers to a polypeptide that comprises an amino acid sequence of a second polypeptide that has been altered by the introduction of at least one amino acid residue substitution, deletion or addition.
  • the derivative polypeptide possesses a similar or identical function as the second polypeptide.
  • “Fragment” refers to a peptide or polypeptide comprising an amino acid sequence of at least 5 amino acid residues (preferably, at least 10 amino acid residues, at least 15 amino acid residues, at least 20 amino acid residues, at least 25 amino acid residues, at least 40 amino acid residues, at least 50 amino acid residues, at least 60 amino residues, at least 70 amino acid residues, at least 80 amino acid residues, at least 90 amino acid residues, at least 100 amino acid residues, at least 125 amino acid residues, at least 150 amino acid residues, at least 175 amino acid residues, at least 200 amino acid residues, or at least 250 amino acid residues) of the amino acid sequence of a second polypeptide.
  • the fragment of a MSPI possesses the functional activity of the MSPI.
  • the "percent identity" of two amino acid sequences or of two nucleic acid sequences can be or is generally determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in either sequences for best alignment with the other sequence) and comparing the amino acid residues or nucleotides at corresponding positions.
  • the "best alignment” is an alignment of two sequences that results in the highest percent identity.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm known to those of skill in the art.
  • An example of a mathematical algorithm for comparing two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877.
  • the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410 have incorporated such an algorithm.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.).
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • Diagnosis refers to diagnosis, prognosis, monitoring, characterizing, selecting patients, including participants in clinical trials, and identifying patients at risk for or having a particular disorder or those most likely to respond to a particular therapeutic treatment, or for assessing or monitoring a patient's response to a particular therapeutic treatment.
  • Treatment refers to therapy, prevention and prophylaxis and particularly refers to the administration of medicine or the performance of medical procedures with respect to a patient, for either prophylaxis (prevention) or to cure the infirmity or malady in the instance where the patient is afflicted.
  • Agent refers to all materials that may be used to prepare pharmaceutical and diagnostic compositions, or that may be compounds, agonists, antagonists, nucleic acids, polypeptides, fragments, isoforms, variants, or other materials that may be used independently for such purposes, all in accordance with the present invention.
  • Highly stringent conditions refers to hybridization to filter-bound DNA in 0.5 M NaHPO4, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65°C, and washing in 0.1xSSC/0.1% SDS at 68°C (Ausubel F.M. et al., eds., 1989, Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc., and John Wiley & Sons, Inc., New York, at p. 2.10.3; incorporated herein by reference in its entirety.)
  • hybridizes For some applications, less stringent conditions for duplex formation are required.
  • moderately stringent conditions refers to washing in 0.2xSSC/0.1% SDS at 42°C (Ausubel et al., 1989, supra).
  • CSF Cerebrospinal fluid
  • “Serum” refers to the supernatant fluid produced by clotting and centrifugal sedimentation of a blood sample.
  • Pulsma refers to the supernatant fluid produced by inhibition of clotting (for example, by citrate or EDTA) and centrifugal sedimentation of a blood sample.
  • Blood as used herein includes serum and plasma.
  • Two-dimensional electrophoresis (2D-electrophoresis) means a technique comprising isoelectric focusing, followed by denaturing electrophoresis; this generates a two-dimensional gel (2D-gel) containing a plurality of separated proteins.
  • the step of denaturing electrophoresis uses polyacrylamide electrophoresis in the presence of sodium dodecyl sulfate (SDS -PAGE).
  • SDS -PAGE sodium dodecyl sulfate
  • the Preferred Technology described in WO 98/23950 and in U.S. Patent Nos 6,064,654, and 6,278.794, each of which is incorporated herein by reference in its entirety with particular reference to the protocol at pages 23-35 of WO 98/23950.
  • the Preferred Technology provides efficient, computer-assisted methods and apparatus for identifying, selecting and characterising biomolecules (e.g. proteins, including glycoproteins) in a biological sample.
  • a two-dimensional array is generated by separating biomolecules on a two-dimensional gel according to their electrophoretic mobility and isoelectric point.
  • a computer-generated digital profile of the array is generated, representing the identity, apparent molecular weight, isoelectric point, and relative abundance of a plurality of biomolecules detected in the two-dimensional array, thereby permitting computer-mediated comparison of profiles from multiple biological samples, as well as computer aided excision of separated proteins of interest.
  • the Basiji thesis provides a phase-sensitive detection system for discriminating modulated fluorescence from baseline noise due to laser scatter or homogeneous fluorescence, but the scanner can also be operated in a non-phase-sensitive mode.
  • This phase-sensitive detection capability would increase the sensitivity of the instrument by an order of magnitude or more compared to conventional fluorescence imaging systems. The increased sensitivity would reduce the sample-preparation load on the upstream instruments while the enhanced image quality simplifies image analysis downstream in the process.
  • a more highly preferred scanner is a modified version of the above described scanner.
  • the gel is transported through the scanner on a precision lead-screw drive system. This is preferable to laying the glass plate on the belt-driven system that is described in the Basiji thesis, as it provides a reproducible means of accurately transporting the gel past the imaging optics.
  • the gel is secured against three alignment stops that rigidly hold the glass plate in a known position. By doing this in conjunction with the above precision transport system, the absolute position of the gel can be predicted and recorded. This ensures that co-ordinates of each feature on the gel can be determined more accurately and communicated, if desired, to a cutting robot for excision of the feature.
  • the carrier that holds the gel has four integral fluorescent markers for use to correct the image geometry. These markers are a quality control feature that confirms that the scanning has been performed correctly.
  • the optical components of the preferred scanner have been inverted.
  • the laser, mirror, waveguide and other optical components are above the glass plate being scanned.
  • the scanner described in the Basiji thesis has these components underneath.
  • the glass plate is mounted onto the scanner gel side down, so that the optical path remains through the glass plate. By doing this, any particles of gel that may break away from the glass plate will fall onto the base of the instrument rather than into the optics. This does not affect the functionality of the system, but increases its reliability.
  • Still more preferred is a modified version of the preferred scanner, in which the signal output is digitised to the full 16-bit data without any peak saturation or without square root encoding of the signal.
  • a compensation algorithm has also been applied to correct for any variation in detection sensitivity along the path of the scanning beam. This variation is due to anomalies in the optics and differences in collection efficiency across the waveguide.
  • a calibration is performed using a perspex plate with an even fluorescence throughout. The data received from a scan of this plate are used to determine the multiplication factors needed to increase the signal from each pixel level to a target level. These factors are then used in subsequent scans of gels to remove any internal optical variations.
  • two-dimensional electrophoresis is used to analyze CSF from a subject, preferably a living subject, in order to detect or quantify the expression of one or more Multiple Sclerosis-Associated Features (MSFs) for screening, prevention or diagnosis of MS, to determine the prognosis of a subject having MS, to monitor progression of MS, to monitor the effectiveness of MS therapy, for identifying patients most likely to respond to a particular therapeutic treatment, or for drug development.
  • MSFs Multiple Sclerosis-Associated Features
  • a number of samples from subjects having MS and samples from subjects free from MS are separated by two-dimensional electrophoresis, and the fluorescent digital images of the resulting gels are matched to a chosen representative primary master gel image.
  • This process allows any gel feature, characterised by its pi and MW, to be identified and examined on any gel of the study.
  • the amount of protein present in a given feature can be measured in each gel; this feature abundance can be averaged amongst gels from similar samples (e.g. gels from samples from subjects having MS).
  • statistical analyses can be conducted on the thus created sample sets, in order to compare 2 or more sample sets to each other.
  • the MSFs disclosed herein have been identified by comparing CSF samples from subjects having MS against CSF samples from subjects free from MS.
  • Subjects free from MS include subjects with no known disease or condition (normal subjects) and subjects with diseases other than MS.
  • the first group consists of MSFs that are decreased in the CSF of subjects having MS as compared with the CSF of subjects free from MS, where the differential presence is significant.
  • MSFs can be described by apparent molecular weight (MW) and isoelectric point (pi) as provided in Table I.
  • the second group consists of MSFs that are increased in the CSF of subjects having MS as compared with the CSF of subjects free from MS where the differential presence is significant.
  • MSFs can be described by MW and pi as provided in Table II:
  • the signal obtained upon analyzing CSF from subjects having MS relative to the signal obtained upon analyzing CSF from subjects free from MS will depend upon the particular analytical protocol and detection technique that is used. Accordingly, the present invention contemplates that each laboratory will, based on the present description, establish a reference range for each MSF in subjects free from MS according to the analytical protocol and detection technique in use, as is conventional in the diagnostic art.
  • at least one control positive CSF sample from a subject known to have MS or at least one control negative CSF sample from a subject known to be free from MS (and more preferably both positive and negative control samples) are included in each batch of test samples analyzed.
  • the level of expression of a feature is determined relative to a background value, which is defined as the level of signal obtained from a proximal region of the image that (a) is equivalent in area to the particular feature in question; and (b) contains no discernable protein feature.
  • the signal associated with an MSF in the CSF of a subject is normalised with reference to one or more ERFs detected in the same 2D gel.
  • ERFs may readily be determined by comparing different samples using the Preferred Technology. Suitable ERFs include O ut are not limited to) thos described in Table HI.
  • the apparent MW and pi of a given feature or protein isoform will vary to some extent depending on the precise protocol used for each step of the 2D electrophoresis and for landmark matching (as described in section 6.1.8 infra).
  • the terms "MW” and "pi" are defined, respectively, to mean the apparent molecular weight and the apparent isoelectric point of a feature or protein isoform as measured in exact accordance with the Reference Protocol identified in Section 6 below.
  • variation in the measured mean pi of an MSF or MSPI is typically less than 3% and variation in the measured mean MW of an MSF or MSPI is typically less than 5%.
  • calibration experiments should be performed to compare the MW and pi for each MSF or protein isoform as detected (a) by the Reference Protocol and (b) by the deviant protocol.
  • MSFs can be used for detection, diagnosis, or monitoring of MS, or for identifying patients most likely to respond to a specific therapeutic treatment, or for drug development.
  • a first sample of body fluid from a subject e.g., a subject suspected of having MS
  • 2D electrophoresis for quantitative detection of one or more of the MSFs as defined in Table I.
  • a decreased abundance of said one or more of these MSFs in the first sample from the subject relative to a second sample from a subject or subjects free from MS (e.g., a control sample or a previously determined reference range) indicates the presence of MS .
  • a first sample of body fluid from a subject is analyzed by 2D electrophoresis for the quantitative detection of one or more of the MSFs as defined in Table ⁇ .
  • An increased abundance of said one or more MSFs in the first sample from the subject relative to a second sample from a subject or subjects free from MS indicates the presence of MS.
  • a first sample of body fliud from a subject is analyzed by 2D electrophoresis for quantitative detection of (a) one or more MSFs or any combination of them, whose decreased abundance indicates the presence of MS, i.e., the MSFs as defined in Table I; and (b) one or more MSFs or any combination of them, whose increased abundance indicates the presence of MS i.e., MSFs as defined in Table H
  • a first sample of body fluid from a subject is analyzed by 2D electrophoresis for quantitative detection of one or more of the MSFs as defined in Table I or Table II:, wherein the ratio of the one or more MSFs relative to an Expression Reference Feature (ERF) indicates whether MS is present.
  • ERP Expression Reference Feature
  • a decrease in one or more MSF/ERF ratios in a first sample relative to the MSF/ERF ratios in a second sample or a reference range indicates the presence of MS; i.e. the MSFs as defined in Table I are suitable MSFs for this purpose.
  • an increase in one or more MSF/ERF ratios in a first sample relative to the MSF/ERF ratios in a second sample or a reference range indicates the presence of MS; the MSFs as defined in Table II are suitable MSFs for this purpose.
  • a first sample of body fluid from a subject is analyzed by 2D electrophoresis for quantitative detection of (a) one or more MSFs, or any combination of them, whose decreased MSF/ERF ratio(s) in a first sample relative to the MSF/ERF ratio(s) in a second sample indicates the presence of MS, i.e., the MSFs as defined in Table I; (b) one or more MSFs, or any combination of them, whose increased MSF/ERF ratio(s) in a first sample relative to the MSF/ERF ratio(s) in a second sample indicates the presence of MS, i.e., the MSFs as defined in Table JJ.
  • CSF from a subject is analyzed for quantitative detection of a plurality of MSFs.
  • MSPIs 5.4 MULTIPLE SCLEROSIS-ASSOCIATED PROTEIN ISOFORMS
  • a sample of body fluid from a subject is analyzed for quantitative detection of one or more Multiple Sclerosis-Associated Protein Isoforms (MSPIs) for screening or diagnosis of MS, to monitor the effectiveness of MS therapy, for identifying patients most likely to respond to a particular therapeutic treatment or for drug development.
  • MSPIs Multiple Sclerosis-Associated Protein Isoforms
  • a given protein may be expressed as variants that differ in their amino acid composition (e.g. as a result of alternative mRNA or premRNA processing, e.g.
  • Multiple Sclerosis-Associated Protein Isoform refers to a polypeptide that is differentially present in a first sample of body fluid from a subject having MS compared with second sample from a subject free from MS.
  • MSPIs Two groups of MSPIs have been identified by amino acid sequencing of MSFs. MSPIs were isolated, subjected to proteolysis, and analyzed by mass spectrometry using the methods and apparatus of the Preferred Technology. One skilled in the art can identify sequence information from proteins analyzed by mass spectrometry and/or tandem mass spectrometry using various spectral interpretation methods and database searching tools. Examples of some of these methods and tools can be found at the Swiss Institute of Bioinformatics web site at http://www.expasy.com/, and the European Molecular Biology Laboratory web site at http://www.narrador.emblrheidelberg.de/GroupPages/PageLink/peptidesearchpage.html.
  • MSPIs were performed primarily using the SEQUEST search program (Eng et al., 1994, J. Am. Soc. Mass Spectrom. 5:976-989) and the method described in PCT/GBOl/04034.
  • the first group consists of MSPIs that are decreased in the CSF of subjects having MS as compared with the CSF of subjects free from MS, where the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these MSPIs identified by tandem mass spectrometry and database searching as described in the Examples, infra are listed in Table IV in addition to the pis and MWs of these MSPIs.
  • the second group comprises MSPIs that are increased in the CSF of subjects having MS as compared with the CSF of subjects free from MS, where the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these MSPIs identified by tandem mass spectrometry and database searching are listed in Table V in addition to the pis and MWs of these MSPIs.
  • the MSPI is a polypeptide comprising a peptide sequence described for that MSPI (preferably comprising a plurality of, more preferably all of, the peptide sequences described for that MSPI) and has a pi of about the value stated for that MSPI (preferably within 10%, more preferably within 5% still more preferably within 1% of the stated value) and has a MW of about the value stated for that MSPI (preferably within 10%, more preferably within 5%, still more preferably within 1% of the stated value).
  • a first sample of body fluid from a subject is analyzed for quantitative detection of one or more of the MSPIs as defined in Table IV, or any combination of them, wherein a decreased abundance of the MSPI or MSPIs (or any combination of them) in the first sample from the subject relative to the second sample from a subject or subjects free from MS (e.g., a control sample or a previously determined reference range) indicates the presence of MS.
  • a first sample of body fluid from a subject is analyzed for quantitative detection of one or more of the MSPIs as defined in Table V, or any combination of them, wherein an increased abundance of the MSPI or MSPIs (or any combination of them) in first sample from the subject relative to the second sample from a subject or subjects free from MS (e.g., a control sample or a previously determined reference range) indicates the presence of MS.
  • a first sample of body fluid from a subject is analyzed for quantitative detection of (a) one or more MSPIs, or any combination of them, whose decreased abundance indicates the presence of MS, i.e., the MSPIs as defined in Table TV; and (b) one or more MSPIs, or any combination of them, whose increased abundance indicates the presence of MS, i.e., the MSPIs as defined in Table V.
  • a first sample of body fluid from a subject is analyzed for quantitative detection of one or more MSPIs and one or more previously l ⁇ iown biomarkers of MS (e.g., candidate markers such as hypersensitive platelet glutamate receptors (Berk et al. Int Clin Psychopharmacol 1999 14, 199-122)).
  • candidate markers such as hypersensitive platelet glutamate receptors (Berk et al. Int Clin Psychopharmacol 1999 14, 199-122)
  • the abundance of each MSPI and known biomarker relative to a control or reference range indicates whether a subject has MS.
  • ERPIs can be identified by partial amino acid sequencing of ERFs, which are described above, using the methods and apparatus of the Preferred Technology.
  • the partial amino acid sequences of an ERPI, and the known proteins to which it is homologous is presented in Table VI.
  • the MSPIs described herein include isoforms of known proteins where the isoforms were not previously known to be associated with MS.
  • the present invention additionally provides: (a) antibodies that bind to said MSPI, to said fragments, or both to said •MSPI and to said fragments.
  • the MSPI is in an isolated form, as used herein, an MSPI is "isolated" when it is present in a preparation that is substantially free of contaminating proteins, i.e., a preparation in which less than 10% (preferably less than 5%, more preferably less than 1%) of the total protein present is contaminating protein(s).
  • a contaminating protein is a protein or protein isoform having a significantly different pi or MW from those of the isolated MSPI, as determined by 2D electrophoresis.
  • a "significantly different" pi or MW is one that permits the contaminating protein to be resolved from the MSPI on 2D electrophoresis, performed according to the Reference Protocol.
  • an isolated polypeptide comprising a peptide with the amino acid sequence identified in Table IV or V for an MSPI, said polypeptide having a pi and MW within 10% (preferably within 5%, more preferably within 1%) of the values identified in Table IV or V for that MSPI.
  • the MSPIs of the invention can be qualitatively or quantitatively detected by any method known to those skilled in the art, including but not limited to the Preferred Technology described herein, kinase assays, enzyme assays, binding assays and other functional assays, irnmunoassays, and western blotting.
  • the MSPIs are separated on a 2-D gel by virtue of their MWs and pis and visualized by staining the gel.
  • the MSPIs are stained with a fluorescent dye and imaged with a fluorescence scanner. Sypro Red (Molecular Probes, Inc., Eugene, Oregon) is a suitable dye for this purpose.
  • a preferred fluorescent dye is Pyridinium, 4-[2-[4-(dipentylamino)-2-trifluoromethylphenyl] ethenyl]-l-(sulfobutyl)-, inner salt. See U.S. Application No. 09/412,168, filed on October 5, 1999, which is incorporated herein by reference in its entirety.
  • MSPIs can be detected in an immunoassay.
  • an immunoassay is performed by contacting a first sample from a subject to be tested with a capture reagent (e.g an antibody) under conditions such that immunospecific binding can occur if the MSPI is present, and detecting or measuring the amount of any immunospecific binding by the capture reagent.
  • a capture reagent e.g an antibody
  • Anti-MSPI antibodies can be produced by the methods and techniques taught herein; examples of such antibodies known in the art are set forth in Table VU. These antibodies shown in Table VU are already known to bind to the protein of which the MSPI is itself a family member.
  • the anti-MSPI antibody preferentially binds to the MSPI rather than to other isoforms of the same protein.
  • the anti-MSPI antibody binds to the MSPI with at least 2-fold greater affinity, more preferably at least 5-fold greater affinity, still more preferably at least 10-fold greater affinity, than to said other isoforms of the same protein.
  • MSPIs can be transferred from a gel to a suitable membrane (e.g. a PVDF membrane) and subsequently probed in suitable assays that include, without limitation, competitive and non-competitive assay systems using techniques such as western blots and "sandwich" irnmunoassays using anti-MSPI antibodies as described herein, e.g., the antibodies raised against the MSPIs of interest.
  • suitable assays include, without limitation, competitive and non-competitive assay systems using techniques such as western blots and "sandwich" irnmunoassays using anti-MSPI antibodies as described herein, e.g., the antibodies raised against the MSPIs of interest.
  • the immunoblots can be used to identify those anti-MSPI antibodies displaying the selectivity required to immuno-specifically differentiate an MSPI from other isoforms encoded by the same gene.
  • an "aberrant level” means a level that is increased or decreased in a first sample compared with the level in a second sample from a subject free from MS or a reference level.
  • binding of antibody in tissue sections can be used to detect aberrant MSPI localization or an aberrant level of one or more MSPIs.
  • antibody to an MSPI can be used to assay a first tissue sample (e.g., a brain biopsy) from a subject for the level of the MSPI where an aberrant level of MSPI is indicative of MS.
  • any suitable immunoassay can be used, including, without limitation, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” irnmunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent irnmunoassays and protein A irnmunoassays.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” irnmunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination
  • an MSPI can be detected in a fluid sample (e.g., CSF, blood, urine, or tissue homogenate) by means of a two-step sandwich assay.
  • a capture reagent e.g., an anti-MSPI antibody
  • the capture reagent can optionally be immobilized on a solid phase.
  • a directly or indirectly labeled detection reagent is used to detect the captured MSPI.
  • the detection reagent is a lectin.
  • any lectin can be used for this purpose that preferentially binds to the MSPI rather than to other isoforms that have the same core protein as the MSPI or to other polypeptides that share the antigenic determinant recognized by the antibody.
  • the chosen lectin binds to the MSPI with at least 2-fold greater affinity, more preferably at least 5-fold greater affinity, still more preferably at least 10-fold greater affinity, than to said other isoforms that have the same core protein as the MSPI or to said other polypeptides that share the antigenic determinant recognized by the antibody.
  • a lectin that is suitable for detecting a given MSPI can readily be identified by methods well known in the art, for instance upon testing one or more lectins enumerated in Table I on pages 158-159 of Sumar et al, Lectins as Indicators of Disease-Associated Glycoforms, In: Gabius H-J & Gabius S (eds.), 1993, Lectins and Glycobiology, at pp. 158-174 (which is incorporated herein by reference in its entirety).
  • Lectins with the desired oligosaccharide specificity can be identified, for example, by their ability to detect the MSPI in a 2D gel, in a replica of a 2D gel following transfer to a suitable solid substrate such as a nitrocellulose membrane, or in a two-step assay following capture by capture reagent.
  • the detection reagent is an antibody, e.g., an antibody that immunospecifically detects post-translational modifications, such as an antibody that immunospecifically binds to phosphorylated amino acids.
  • antibodies examples include those that bind to phosphotyrosine (BD Transduction Laboratories, 2002, catalog nos.:P11120; P39020), those that bind to phosphoserine (Zymed Laboratories Inc. 2002, South San Francisco, CA, catalog no. 61-8100) and those that bind to phosphothreonine (Zymed Laboratories Inc., 2002, South San Francisco, CA, catalog nos. 71-8200, 13-9200).
  • phosphotyrosine BD Transduction Laboratories, 2002, catalog nos.:P11120; P39020
  • those that bind to phosphoserine Zymed Laboratories Inc. 2002, South San Francisco, CA, catalog no. 61-8100
  • phosphothreonine Zymed Laboratories Inc., 2002, South San Francisco, CA, catalog nos. 71-8200, 13-9200.
  • a gene encoding an MSPI, a related gene, or related nucleic acid sequences or subsequences, including complementary sequences can also be used in hybridization assays.
  • a nucleotide encoding an MSPI, or subsequences thereof comprising at least 8 nucleotides, preferably at least 12 nucleotides, and most preferably at least 15 nucleotides can be used as a hybridization probe.
  • Hybridization assays can be used for detection, prognosis, diagnosis, or monitoring of conditions, disorders, or disease states, associated with aberrant expression of genes encoding MSPIs, or for differential diagnosis of subjects with signs or symptoms suggestive of MS.
  • such a hybridization assay can be carried out by a method comprising contacting a subject's sample containing nucleic acid with a nucleic acid probe capable of hybridizing to a DNA or RNA that encodes an MSPI, under conditions such that hybridization can occur, and detecting or measuring any resulting hybridization.
  • Nucleotides can be used for therapy of subjects having MS, as described below.
  • kits comprising an anti-MSPI antibody.
  • a kit may optionally comprise one or more of the following: (1) instructions for using the anti-MSPI antibody for diagnosis, prognosis, therapeutic monitoring or any combination of these applications; (2) a labeled binding partner to the antibody; (3) a solid phase (such as a reagent strip) upon which the anti-MSPI antibody is immobilized; and (4) a label or insert indicating regulatory approval for diagnostic, prognostic or therapeutic use or any combination thereof.
  • the anti-MSPI antibody itself can be labeled with a detectable marker, e.g., a chemiluminescent, enzymatic, fluorescent, or radioactive moiety.
  • kits comprising a nucleic acid probe capable of hybridizing to RNA encoding an MSPI.
  • a kit comprises in one or more containers a pair of primers (e.g., each in the size range of 6-30 nucleotides, more preferably 10-30 nucleotides and still more preferably 10-20 nucleotides) that under appropriate reaction conditions can prime amplification of at least a portion of a nucleic acid encoding an MSPI, such as by polymerase chain reaction (see, e.g., Innis et al., 1990, PCR Protocols, Academic Press, Inc., San Diego, CA), ligase chain reaction (see EP 320,308) use of Q replicase, cyclic probe reaction, or other methods known in the art.
  • primers e.g., each in the size range of 6-30 nucleotides, more preferably 10-30 nucleotides and still more preferably 10-20 nucleotides
  • Kits are also provided which allow for the detection of a plurality of MSPIs or a plurality of nucleic acids each encoding an MSPI.
  • a kit can optionally further comprise a predetermined amount of an isolated MSPI protein or a nucleic acid encoding an MSPI, e.g., for use as a standard or control.
  • the uni-variate differential analysis tools are useful in identifying individual MSFs or MSPIs that are diagnostically associated with MS or in identifying individual MSPIs that regulate the disease process.
  • the disease process is associated with a combination of MSFs or MSPIs (and to be regulated by a combination of MSPIs), rather than individual MSFs and MSPIs in isolation.
  • the strategies for discovering such combinations of MSFs and MSPIs differ from those for discovering individual MSFs and MSPIs. In such cases, each individual MSF and MSPIs can be regarded as one variable and the disease can be regarded as a joint, multi-variate effect caused by interaction of these variables.
  • the first step is to identify a collection of MSFs or MSPIs that individually show a significant aberrant expression in MS.
  • the association between the identified MSFs or MSPIs and MS need not be as highly significant as is desirable when an individual MSF or MSPI is used as a diagnostic. Any of the tests discussed above (fold changes, Wilcoxon rank sum test, etc.) can be used at this stage.
  • a sophisticated multi-variate analysis capable of identifying clusters can then be used to estimate the significant multivariate associations with MS.
  • LDA Linear Discriminant Analysis
  • MSFs or MSPIs variables
  • MSPIs Linear Discriminant Analysis
  • a set of weights is associated with each variable (i.e., MSF or MSPI) so that the linear combination of weights and the measured values of the variables can identify the disease state by discriminating between subjects having MS and subjects free from MS.
  • Enhancements to the LDA allow stepwise inclusion (or removal) of variables to optimize the discriminant power of the model.
  • the result of the LDA is therefore a cluster of MSFs or MSPIs which can be used, without limitation, for diagnosis, prognosis, therapy or drug development.
  • LDA Flexible Discriminant Analysis
  • results of the discriminant analysis can be verified by post-hoc tests and also by repeating the analysis using alternative techniques such as classification trees.
  • a further category of MSFs or MSPIs can be identified by qualitative measures by comparing the percentage feature presence of an MSF or MSPI of a first sample or sample set (e.g., samples from diseased subjects) with the percentage feature presence of an MSF or MSPI in a second sample or sample set (e.g., samples from control subjects).
  • the "percentage feature presence" of an MSF or MSPI is the percentage of samples in a sample set in which the MSF or MSPI is detectable by the detection method of choice. For example, if an MSF is detectable in 95 percent of samples from diseased subjects, the percentage feature presence of that MSF in that sample set is 95 percent. If only 5 percent of samples from non-diseased subjects have detectable levels of the same MSF, detection of that MSF in the sample of a subject would suggest that it is likely that the subject suffers from MS.
  • the diagnostic methods and compositions of the present invention can assist in monitoring a clinical study, e.g. to evaluate drugs for therapy of MS.
  • candidate molecules are tested for their ability to restore MSF or MSPI levels in a subjecthaving MS to levels found in subjects free from MS or, in a treated subject (e.g. after treatment for relapsing-remitting MS with include Interferon ⁇ -lb (BetaseronTM, BetaferonTM), Interferon ⁇ -la (AvonexTM, RebifTM), Glatiramer acetate (CopaxoneTM), intravenous immunoglobulin and, for incidences of acute relapse, therapies with corticosteroids (Noseworthy (1999) Nature 399:suppl. A40-A47), to preserve MSF or MSPI levels at or near non-MS values.
  • the levels of one or more MSFs or MSPIs can be assayed.
  • the methods and compositions of the present invention are used to screen candidates for a clinical study to identify individuals having MS; such individuals can then be either excluded from or included in the study or can be placed in a separate cohort for treatment or analysis. If desired, the candidates can concurrently be screened to identify individuals with MS; procedures for these screens are well known in the art.
  • the invention provides isolated mammalian MSPI, preferably human MSPI, and fragments thereof which comprise an antigenic determinant (i.e., can be recognized by an antibody) or which are otherwise functionally active, as well as nucleic acid sequences encoding the foregoing.
  • "Functionally active” as used herein refers to material displaying one or more functional activities associated with a full-length (wild-type) MSPI, e.g., binding to an MSPI substrate or MSPI binding partner, antigenicity (binding to an anti-MSPI antibody), immunogenicity, enzymatic activity and the like.
  • the invention provides fragments of an MSPI comprising at least 5 amino acids, at least 10 amino acids, at least 50 amino acids, or at least 75 amino acids. Fragments lacking some or all of the regions of an MSPI are also provided, as are polypeptides (e.g., fusion proteins) comprising such fragments. Nucleic acids encoding the foregoing are provided.
  • the gene. product can be analyzed. This is achieved by assays based on the physical or functional properties of the product, including radioactive labeling of the product followed by analysis by gel electrophoresis, immunoassay, etc.
  • the MSPIs identified herein can be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • the entire amino acid sequence of the MSPI can be deduced from the nucleotide sequence of the gene coding region contained in the recombinant nucleic acid.
  • the protein can be synthesized by standard chemical methods known in the art (e.g., see Hunkapiller et al, 1984, Nature 310:105-111).
  • native MSPIs can be purified from natural sources, by standard methods such as those described above (e.g., immunoaffinity purification).
  • MSPIs are isolated by the Preferred Technology described supra.
  • a narrow-range "zoom gel” having a pH range of 2 pH units or less is preferred for the isoelectric step, according to the method described in Westermeier, 1993, Electrophoresis in Practice (VCH, Weinheim, Germany), pp. 197-209 (which is incorporated herein by reference in its entirety); this modification permits a larger quantity of a target protein to be loaded onto the gel, and thereby increases the quantity of isolated MSPIs that can be recovered from the gel.
  • the Preferred Technology typically provides up to 100 ng, and can provide up to 1000 ng, of an isolated MSPI in a single run.
  • a zoom gel can be used in any separation strategy which employs gel isoelectric focusing.
  • the invention thus provides an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein; any of the foregoing can be produced by recombinant DNA techniques or by chemical synthetic methods.
  • nucleotide sequences of the present invention including DNA and RNA, and comprising a sequence encoding an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein, may be synthesized using methods known in the art, such as using conventional chemical approaches or polymerase chain reaction (PCR) amplification.
  • the nucleotide sequences of the present invention also permit the identification and cloning of the gene encoding an MSPI homolog or MSPI ortholog including, for example, by screening cDNA libraries, genomic libraries or expression libraries.
  • oligonucleotides can be designed for all MSPI peptide fragments identified as part of the same protein.
  • PCR reactions under a variety of conditions can be performed with relevant cDNA and genomic DNAs (e.g., from CSF or from cells of the immune system) from one or more species.
  • vectorette reactions can be performed on any available cDNA and genomic DNA using the oligonucleotides (which preferably are nested) as above.
  • Vectorette PCR is a method that enables the amplification of specific DNA fragments in situations where the sequence of only one primer is known.
  • Vectorette PCR may be performed with probes that are, for example, anchored degenerate oligonucleotides (or most likely oligonucleotides) coding for MSPI peptide fragments, using as a template a genomic library or cDNA library pools.
  • Anchored degenerate oligonucleotides (and most likely oligonucleotides) can be designed for all MSPI peptide fragments.
  • oligonucleotides may be labeled and hybridized to filters containing cDNA and genomic DNA libraries. Oligonucleotides to different peptides from the same protein will often identify the same members of the library.
  • the cDNA and genomic DNA libraries may be obtained from any suitable or desired mammalian species, for example from humans.
  • Nucleotide sequences comprising a nucleotide sequence encoding MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein of the present invention are useful for their ability to hybridize selectively with complementary stretches of genes encoding other proteins.
  • a variety of hybridization conditions may be employed to obtain nucleotide sequences at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% identical, or 100% identical, to the sequence . of a nucleotide encoding an MSPI.
  • hybridization conditions For a high degree of selectivity, relatively stringent conditions are used to form the duplexes, such as low salt or high temperature conditions. Hybridization conditions can also be rendered more stringent by the addition of increasing amounts of formamide, to destabilize the hybrid duplex. Thus, particular hybridization conditions can be readily manipulated, and will generally be chosen depending on the desired results. In general, convenient hybridization temperatures in the presence of 50% formamide are: 42°C for a probe which is 95 to 100% identical to the fragment of a gene encoding an MSPI, 37°C for 90 to 95% identity and 32°C for 70 to 90% identity.
  • DNA fragments are generated, some of which will encode parts or the whole of an MSPI.
  • Any suitable method for preparing DNA fragments may be used in the present invention.
  • the DNA may be cleaved at specific sites using various restriction enzymes.
  • the DNA fragments can then be separated according to size by standard techniques, including but not limited to agarose and polyacrylamide gel electrophoresis, column chromatography and sucrose gradient centrifugation.
  • the DNA fragments can then be inserted into suitable vectors, including but not limited to plasmids, cosmids, bacteriophages lambda or T4, and yeast artificial chromosome (YAC).
  • suitable vectors including but not limited to plasmids, cosmids, bacteriophages lambda or T4, and yeast artificial chromosome (YAC).
  • YAC yeast artificial chromosome
  • the genomic library may be screened by nucleic acid hybridization to labeled probe (Benton and Davis, 1977, Science 196:180; Grunstein and Hogness, 1975, Proc. Natl. Acad. Sci. U.S.A. 72:3961).
  • the genomic libraries may be screened with labeled degenerate oligonucleotide probes corresponding to the amino acid sequence of any peptide of the MSPI using optimal approaches well known in the art.
  • Any probe used is at least 10 nucleotides, at least 15 nucleotides, at least 20 nucleotides, at least 25 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, or at least 100 nucleotides.
  • MSPIs disclosed herein were found to correspond to isoforms of previously identified proteins encoded by genes whose sequences are publicly known. (Sequence analysis and protein identification of MSPIs was carried out using the methods described in Section 6.1.14). To screen such a gene, any probe may be used that is complementary to the gene or its complement; preferably the probe is 10 nucleotides or longer, more preferably 15 nucleotides or longer.
  • SWISS-PROT and trEMBL databases (held by the Swiss Institute of Bioinformatics (SIB) and the European Bioinformatics Institue (EBI) which are available at http://www.expasy.com/) and the GenBank database (held by the National Institute of Health (NIH) which is available at http://www.ncbi.nlm.nih.gov/GenBank/) provide protein sequences for the MSPIs listed in Tables IV and V under the following accession numbers and each sequence is incorporated herein by reference:
  • degenerate probes or probes taken from the sequences described above by accession number may be used for screening.
  • they can be constructed from the partial amino sequence information obtained from tandem mass spectra of tryptic digest peptides of the MSPI.
  • any probe may be used that is complementary to the gene or its complement; the probe is 10 nucleotides or longer, preferably 15 nucleotides or longer.
  • Hybridization of such oligonucleotide probes to genomic libraries is carried out using methods known in the art. For example, hybridization with one of the above-mentioned degenerate sets of oligonucleotide probes, or their complement (or with any member of such a set, or its complement) can be performed under highly stringent or moderately stringent conditions as defined supra, or can be carried out in 2X SSC, 1.0% SDS at 50°C and washed using the washing conditions described supra for highly stringent or moderately stringent hybridization.
  • clones containing nucleotide sequences encoding the MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein any of the foregoing may also be obtained by screening expression libraries.
  • DNA from the relevant source is isolated and random fragments are prepared and ligated into an expression vector (e.g., a bacteriophage, plasmid, phagemid or cosmid) such that the inserted sequence in the vector is capable of being expressed by the host cell into which the vector is then introduced.
  • an expression vector e.g., a bacteriophage, plasmid, phagemid or cosmid
  • Various screening assays can then be used to select for the expressed MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein.
  • the various anti-MSPI antibodies of the invention can be used to identify the desired clones using methods known in the art. See, for example, Harlow and Lane, 1988, Antibodies: A
  • colonies or plaques containing DNA that encodes MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein can be detected using DYNA Beads according to Olsvick et al., 29fh ICAAC, Houston, Tex. 1989, incorporated herein by reference.
  • Anti-MSPI antibodies are crosslinked to tosylated DYNA Beads M280, and these antibody-containing beads are then contacted with colonies or plaques expressing recombinant polypeptides. Colonies or plaques expressing an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein are identified as any of those that bind the beads.
  • the anti-MSPI antibodies can be nonspecifically immobilized to a suitable support, such as silica or Celite7 resin. This material is then used to adsorb to bacterial colonies expressing the MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein as described herein.
  • PCR amplification may be used to isolate from genomic DNA a substantially pure DNA (i.e., a DNA substantially free of contaminating nucleic acids) encoding the entire MSPI or a part thereof.
  • a substantially pure DNA i.e., a DNA substantially free of contaminating nucleic acids
  • a DNA is at least 95% pure, more preferably at least 99% pure.
  • Oligonucleotide sequences, degenerate or otherwise, that correspond to peptide sequences of MSPIs disclosed herein can be used as primers.
  • PCR can be carried out, e.g., by use of a Perkin-Elmer Cetus thermal cycler and Taq polymerase (Gene AmpTM or AmpliTaqTM DNA polymerase).
  • a Perkin-Elmer Cetus thermal cycler and Taq polymerase Gene AmpTM or AmpliTaqTM DNA polymerase.
  • One can choose to synthesize several different degenerate primers, for use in the PCR reactions. It is also possible to vary the stringency of hybridization conditions used in priming the PCR reactions, to allow for greater or lesser degrees of nucleotide sequence similarity between the degenerate primers and the corresponding sequences in the DNA.
  • an MSPI After successful amplification of a segment of the sequence encoding an MSPI, that segment may be molecularly cloned and sequenced, and utilized as a probe to isolate a complete genomic clone. This, in turn, will permit the determination of the gene's complete nucleotide sequence, the analysis of its expression, and the production of its protein product for functional analysis, as described infra.
  • the gene encoding an MSPI can also be identified by mRNA selection by nucleic acid hybridization followed by in vitro translation. In this procedure, fragments are used to isolate complementary mRNAs by hybridization. Such DNA fragments may represent available, purified DNA encoding an MSPI of another species (e.g., mouse, human).
  • Immunoprecipitation analysis or functional assays e.g., aggregation ability in vitro; binding to receptor
  • specific mRNAs may be selected by adsorption of polysomes isolated from cells to immobilized antibodies that specifically recognize an MSPI.
  • a radiolabeled cDNA encoding an MSPI can be synthesized using the selected mRNA (from the adsorbed polysomes) as a template. The radiolabeled mRNA or cDNA may then be used as a probe to identify the DNA fragments encoding an MSPI from among other genomic DNA fragments.
  • RNA for cDNA cloning of the gene encoding an MSPI can be isolated from cells which express the MSPI.
  • Any suitable eukaryotic cell can serve as the nucleic acid source for the molecular cloning of the gene encoding an MSPI.
  • the nucleic acid sequences encoding the MSPI can be isolated from vertebrate, mammalian, primate, human, porcine, bovine, feline, avian, equine, canine or murine sources.
  • the DNA may be obtained by standard procedures known in the art from cloned DNA (e.g., a DNA "library”), by chemical synthesis, by cDNA cloning, or by the cloning of genomic DNA, or fragments thereof, purified from the desired cell.
  • Clones derived from genomic DNA may contain regulatory and intron DNA regions in addition to coding regions; clones derived from cDNA will contain only exon sequences.
  • the identified and isolated gene or cDNA can then be inserted into any suitable cloning vector.
  • vector-host systems known in the art may be used.
  • the vector system chosen be compatible with the host cell used.
  • Such vectors include, but are not limited to, bacteriophages such as lambda derivatives, plasmids such as PBR322 or pUC plasmid derivatives or the
  • adenoviruses adeno-associated viruses or retroviruses.
  • the insertion into a cloning vector can be accomplished, for example, by ligating the DNA fragment into a cloning vector which has complementary cohesive termini. However, if the complementary restriction sites used to fragment the DNA are not present in the cloning vector, the ends of the DNA molecules may be enzymatically modified. Alternatively, any site desired may be produced by ligating nucleotide sequences (linkers) onto the DNA termini; these ligated linkers may comprise specific chemically synthesized oligonucleotides encoding restriction endonuclease recognition sequences.
  • linkers may comprise specific chemically synthesized oligonucleotides encoding restriction endonuclease recognition sequences.
  • the cleaved vector and the gene encoding an MSPI may be modified by homopolymeric tailing.
  • Recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, etc., so that many copies of the gene sequence are generated.
  • transformation of host cells with recombinant DNA molecules that incorporate the isolated gene encoding the MSPI, cDNA, or synthesized DNA sequence enables generation of multiple copies of the gene.
  • the gene may be obtained in large quantities by growing transformants, isolating the recombinant DNA molecules from the transformants and, when necessary, retrieving the inserted gene from the isolated recombinant DNA.
  • nucleotide sequences of the present invention include nucleotide sequences encoding amino acid sequences with substantially the same amino acid sequences as native MSPI, nucleotide sequences encoding amino acid sequences with functionally equivalent amino acids, nucleotide sequences encoding MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein.
  • an isolated nucleic acid molecule encoding an MSPI-related polypeptide can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of an MSPI such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
  • Standard techniques known to those of skill in the art can be used to introduce mutations, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis.
  • conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a side chain with a similar charge.
  • Families of amino acid residues having side chains with similar charges have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed and the activity of the protein can be determined.
  • the nucleotide sequence coding for an MSPI, MSPI fragment or MSPI-related polypeptide or other derivative of any of the foregoing can be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted protein-coding sequence.
  • the necessary transcriptional and translational signals can also be supplied by the native gene encoding the MSPI or its flanking regions, or the native gene encoding the MSPI-related polypeptide or its flanking regions.
  • a variety of host- vector systems may be utilized in the present invention to express the protein-coding sequence.
  • mammalian cell systems infected with virus e.g., vaccinia virus, adenovirus, etc.
  • insect cell systems infected with virus e.g., baculovirus
  • microorganisms such as yeast containing yeast vectors; or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA.
  • the expression elements of vectors vary in their strengths and specificities. Depending on the host-vector system utilized, any one of a number of suitable transcription and translation elements may be used.
  • a nucleotide sequence encoding a human gene or a nucleotide sequence encoding a functionally active portion of a human MSPI
  • a fragment of an MSPI comprising a domain of the MSPI is expressed.
  • any of the methods previously described for the insertion of DNA fragments into a vector may be used to construct expression vectors containing a chimeric gene consisting of appropriate transcriptional and translational control signals and the protein coding sequences. These methods may include in vitro recombinant DNA and synthetic techniques and in vivo recombinants (genetic recombination). Expression of nucleic acid sequence encoding an MSPI or fragment thereof may be regulated by a second nucleic acid sequence so that the MSPI or fragment is expressed in a host transformed with the recombinant DNA molecule. For example, expression of an MSPI may be controlled by any promoter or enhancer element known in the art.
  • Promoters which may be used to control the expression of the gene encoding an MSPI or an MSPI-related polypeptide include, but are not limited to, the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Cell 22:787-797), the herpes thymidine kinase promoter (Wagner et al, 1981, Proc. Natl. Acad. Sci. U.S.A.
  • promoter of the photosynfhetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al., 1984, Nature 310:115-120); promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter, and the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gehe control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639-646; Ornitz et al., 1986, Cold Spring Harbor Symp.
  • alpha.1-antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel. 1:161-171), beta-globin gene control region which is active in myeloid cells (Mogram et al, 1985, Nature 315:338-340; Kollias et al., 1986, Cell 46:89-94; myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712); myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286); neuronal-specific enolase (NSE) which is active in neuronal cells (Morelli et al., 1999, Gen.
  • NSE neuronal-specific enolase
  • BDNF brain-derived neurotrophic factor
  • GFAP glial fibrillary acidic protein
  • a vector in a specific embodiment, comprises a promoter operably linked to an MSPI-encoding nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
  • an expression construct is made by subcloning an MSPI, MSPI fragment or MSPI-related polypeptide coding sequence into the EcoRI restriction site of each of the three pGEX vectors (Glutathione S-Transferase expression vectors; Smith and Johnson, 1988, Gene 7:31-40). This allows for the expression of the MSPI product or MSPI- related polypeptide from the subclone in the correct reading frame.
  • the MSPI coding sequence or MSPI-related polypeptide coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo . recombination.
  • Insertion in a non-essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:355-359).
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., 1987, Methods in Enzymol. 153:51-544).
  • Expression vectors containing inserts of a gene encoding an MSPI, MSPI fragment or MSPI-related polypeptide can be identified by three general approaches: (a) nucleic acid hybridization, (b) presence or absence of "marker" gene functions, and (c) expression of inserted sequences.
  • the presence of a gene encoding an MSPI inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted gene encoding an MSPI.
  • the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.) caused by the insertion of a gene encoding an MSPI in the vector.
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.
  • recombinant expression vectors can be identified by assaying the gene product (i.e., MSPI) expressed by the recombinant.
  • assays can be based, for example, on the physical or functional properties of the MSPI in in vitro assay systems, e.g., binding with anti-MSPI antibody.
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus, expression of the genetically engineered MSPI or MSPI-related polypeptide may be controlled.
  • different host cells have characteristic and specific mechanisms for the translational and post-translational processing and modification (e.g., glycosylation, phosphorylation of proteins).
  • Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. For example, expression in a bacterial system will produce an unglycosylated product and expression.in yeast will produce a glycosylated product.
  • Eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, HEK293, 3T3, WI38, and in particular, neuronal cell lines such as, for example, SK-N-AS, SK-N-FI, SK-N-DZ human neuroblastomas (Sugimoto et al., 1984, J. Natl. Cancer Inst. 73: 51-57), SK-N-SH human neuroblastoma (Biochim. Biophys. Acta, 1982, 704: 450-460), Daoy human cerebellar medulloblastoma (He et al., 1992, Cancer Res.
  • SK-N-AS SK-N-FI
  • SK-N-DZ human neuroblastomas Sugimoto et al., 1984, J. Natl. Cancer Inst. 73: 51-57
  • SK-N-SH human neuroblastoma Biochim. Biophys. Acta, 1982
  • DBTRG-05MG glioblastoma cells (Kruse et al., 1992, In Vitro Cell. Dev. Biol. 28A: 609-6.14), IMR-32 human neuroblastoma (Cancer Res., 1970, 30: 2110-2118), 1321N1 human astrocytoma (Proc. Natl Acad. Sci. USA ,1977, 74: 4816), MOG-G-CCM human astrocytoma (Br. J. Cancer, 1984, 49: 269), U87MG human glioblastoma-astrocytoma (Acta Pathol. Microbiol.
  • cell lines which stably express the MSPI may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched medium, and then are switched to a selective medium.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the MSPI.
  • Such engineered cell lines may be particularly . . useful in screening and evaluation of agents that affect the endogenous activity of the MSPI.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler, et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. Sci. USA .
  • adenine phosphoribosyltransferase genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler, et al., . 1980, Natl. Acad. Sci. USA 77:3567; O'Hare, et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc.
  • the MSPI, fragment, analog, or derivative may be expressed as a fusion, or chimeric protein product (comprising the protein, fragment, analog, or derivative joined via a peptide bond to a heterologous protein sequence).
  • the polypeptides of the present invention may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any . combination thereof and portions thereof) resulting in chimeric polypeptides.
  • immunoglobulins IgA, IgE, IgG, IgM
  • CHI constant domain of immunoglobulins
  • CH2, CH3, or any . combination thereof and portions thereof resulting in chimeric polypeptides.
  • Such fusion proteins may facilitate purification, increase half-life in vivo, and enhance the delivery of an antigen across an epithelial barrier to the immune system.
  • Nucleic acids encoding an MSPI, a fragment of an MSPI, an MSPI-related polypeptide, or a fragment of an MSPI-related polypeptide can fused to an epitope tag (e.g., the hemagglutinin ("HA") tag or flag tag) to aid in detection and purification of the expressed polypeptide.
  • an epitope tag e.g., the hemagglutinin ("HA") tag or flag tag
  • HA hemagglutinin
  • a system described by Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al.,
  • Fusion proteins can be made by ligating the appropriate nucleic acid sequences encoding the desired amino acid sequences to each other by methods known in the art, in the proper coding frame, and expressing the chimeric product by methods commonly known in the art.
  • a fusion protein may be made by protein synthetic techniques, e.g., by use of a peptide synthesizer. Both cDNA and genomic sequences can be cloned and expressed.
  • domains of some MSPIs are known in the art and have been described in the scientific literature. Moreover, domains of an MSPI can be identified using techniques known to those of skill in the art. For example, one or more domains of an MSPI can be identified by using one or more of the following programs: ProDom, TMpred, and SAPS.
  • ProDom compares the amino acid sequence of a polypeptide to a database of compiled domains (see, e.g., http://www.toulouse.inra.fr/prodom.html; Corpet F., Gouzy J. & Kahn D., 1999, Nucleic Acids Res., 27:263-267).
  • TMpred predicts membrane-spanning regions of a polypeptide and their orientation.
  • TMbase a database of naturally occuring transmembrane proteins
  • the SAPS program analyzes polypeptides for statistically significant features like charge-clusters, repeats, hydrophobic regions, compositional domains (see, e.g., Brendel et al.,
  • an MSPI has an amino acid sequence sufficiently similar to an identified domain of a known polypeptide.
  • the term "sufficiently similar” refers to a first amino acid or nucleotide sequence which contains a sufficient number of identical or equivalent (e.g., with a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have or encode a common structural domain or common functional activity or both.
  • an MSPI domain can be assessed for its function using techniques well known to those of skill in the art.
  • a domain can be assessed for its kinase activity or for its ability to bind to DNA using techniques known to the skilled artisan.
  • Kinase activity can be assessed, for example, by measuring the ability of a polypeptide to phosphorylate a substrate.
  • DNA binding activity can be assessed, for example, by measuring the ability of a polypeptide to bind to a DNA binding element in a electromobility shift assay.
  • the function of a domain of an MSPI is determined using an assay described in one or more of the references identified in Table IX, infra.
  • an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein or derivative of any of the foregoing may be used as an immunogen to generate antibodies which immunospecifically bind such an immunogen.
  • immunogens can be isolated by any convenient means, including the methods described above.
  • Antibodies of the invention include, but are not limited to polyclonal, monoclonal, bispecific, humanized or chimeric antibodies, single chain antibodies, Fab fragments and F(ab') fragments, fragments . produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen.
  • the immunoglobulin molecules of the invention can be of any class (e.g., IgG, IgE, IgM, IgD and IgA ) or subclass of immunoglobulin molecule.
  • antibodies that recognize gene products of genes encoding MSPIs are publicly available.
  • antibodies that recognize these MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion proteins include antibodies recognizing MSPI-1, MSPI-2, MSPI-3, MSPI-4, MSPI-5.1, MSPI-6, MSPI-7, MSPI-8.1, MSPI-8.2, MSPI-9, MSPI-10, MSPI-11, MSPI-12.1, MSPI-12.2, MSPI-13, MSPI-14, MSPI-15, MSPI-16, MSPI- 18, MSPI-22, MSPI-24, MSPI-27, MSPI-28, MSPI-29, MSPI-30, MSPI-31, MSPI-32, MSPI- 33.1, MSPI-36, MSPI-37, MSPI-39.1, MSPI-40, MSPI-42.2, MSPI-47, MSPI-48.1, MSPI- 51.1, MSPI-52.1, MSPI-52.2, MSPI-54, MSPI-55, MSPI-56, MSPI-58, MSPI-60, MSPI-65, MSPI-67.1, MSPI-67.2, MSPI-70, MSPL72,
  • MSPI-238 MSPI-239.1, MSPI-239.2, MSPI-240.1, MSPI-240.2, MSPI-241.2, MSPI-242.2, MSPI-243, MSPI-244, MSPI-245, MSPI-246.1, MSPI-246.2, MSPI-248, MSPI-251, MSPI- 257.2, MSPI-257.3, MSPI-260.2, MSPI-265, MSPI-266, MSPI-268.1, MSPI-270, MSPI-273, .
  • antibodies to a specific domain of an MSPI are produced.
  • hydrophilic fragments of an MSPI are used as immunogens for antibody production.
  • screening for the desired antibody can be accomplished by techniques known in the art, e.g. ELISA (enzyme-linked immunosorbent assay).
  • ELISA enzyme-linked immunosorbent assay
  • an antibody that specifically binds a first MSPI homolog but which does not specifically bind to (or binds less avidly to) a second MSPI homolog one can select on the basis of positive binding to the first MSPI homolog and a lack of binding to (or reduced binding to) the second MSPI homolog.
  • a different isoform of the same protein such as a different glycoform having the same core peptide as the MSPI
  • the present invention provides an antibody (preferably a monoclonal antibody) that binds with greater affinity (preferably at least 2-fold, more preferably at least 5-fold still more preferably at least 10-fold greater affinity) to an MSPI than to a different isoform or isoforms (e.g., glycoforms) of the MSPI.
  • an antibody preferably a monoclonal antibody
  • binds with greater affinity preferably at least 2-fold, more preferably at least 5-fold still more preferably at least 10-fold greater affinity
  • Polyclonal antibodies which may be used in the methods of the invention are heterogeneous populations of antibody molecules derived from the sera of immunized animals. Unfractionated immune serum can also be used. Various procedures known in the art may be used for the production of polyclonal antibodies to an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein. In a particular embodiment, rabbit polyclonal antibodies to an epitope of an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI- fusion protein can be obtained.
  • various host animals can be immunized by injection with the native or a synthetic (e.g., recombinant) version of an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein, including but not limited to rabbits, mice, rats, etc.
  • a synthetic (e.g., recombinant) version of an MSPI, MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein including but not limited to rabbits, mice, rats, etc.
  • the Preferred ' Technology described herein provides isolated MSPIs suitable for such immunization. If the MSPI is purified by gel electrophoresis, the MSPI can be used for immunization with or without prior extraction from the polyacrylamide gel.
  • adjuvants may be used to enhance the immunological response, depending on the host species, including, but not limited to, complete or incomplete Freund's adjuvant, a mineral gel such as aluminum hydroxide, surface active substance such as lysolecithin, pluronic polyol, a polyanion, a peptide, an oil emulsion, keyhole limpet hemocyanin, dinitrophenol, and an adjuvant such as BCG (bacille Calmette-Guerin) or corynebacterium parvum. Additional adjuvants are also well known in the art.
  • any technique which provides for the production of antibody molecules by continuous cell lines in culture may be used.
  • the hybridoma technique originally developed by Kohler and Milstein (1975, Nature 256:495-497), as well as the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies Colde etal., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof.
  • the hybridoma producing the mAbs of the invention may be cultivated in vitro or in vivo.
  • mAbs can be produced in germ-free animals utilizing known technology (PCT/US90/02545).
  • the mAbs include but are not limited to human mAbs and chimeric mAbs (e.g., human-mouse chimeras).
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a human immunoglobulin constant region and a variable region derived from a murine mAb.
  • Humanized antibodies are antibody molecules from non-human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule.
  • Chimeric and humanized mAbs can be produced by recombinant DNA techniques known in the art, for example using methods described in WO 87/02671; EP 184, 187 A; EP 171.496A; EP 173,494A; WO 86/01533; US 4,816,567; EP 125,023A; Better et al, 1988, Science 240:1041-1043; Liu et al., 1987, Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al., 1987, J. Immunol.
  • Fully human antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Such antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of an MSPI of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope.
  • the antibodies of the present invention can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with, antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene UI or gene VIU protein.
  • Phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in WO 92/22324; Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al, AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988).
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details for generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 1986, 121:210.
  • the invention provides functionally active fragments, derivatives or analogs of the anti-MSPI immunoglobulin molecules.
  • Functionally active means that the fragment, derivative or analog is able to elicit anti-anti-idiotype antibodies (i.e., tertiary antibodies) that recognize the same antigen that is recognized by the antibody from which the fragment, derivative or analog is derived.
  • antigenicity of the idiotype of the immunoglobulin molecule may be enhanced by deletion of framework and CDR sequences that are C-terminal to the CDR sequence that specifically recognizes the antigen.
  • synthetic peptides containing the CDR sequences can be used in binding assays with the antigen by any binding assay method known in the art.
  • the present invention provides antibody fragments such as, but not limited to, F(ab')2 fragments and Fab fragments.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • F(ab')2 fragments consist of the variable region, the light chain constant region and the CHI domain of the heavy chain and are generated by pepsin digestion of the antibody molecule.
  • Fab fragments are generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • the invention also provides heavy chain and light chain dimers of the antibodies of the invention, or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs) (e.g., as described in U.S. 4,946,778; Bird, 1988, Science
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may be used (Skerra et al., 1988, Science 242:1038-1041).
  • the invention provides fusion proteins of the immunoglobulins of the invention (or functionally active fragments thereof), for example in which the immunoglobulin is. fused via a covalent bond (e.g., a peptide bond), at either the N-terminus or the C-terminus to an amino acid sequence of another protein (or portion thereof, preferably at least 10, 20 or 50 amino acid portion of the protein) that is not the immunoglobulin.
  • a covalent bond e.g., a peptide bond
  • the immunoglobulin, or fragment thereof is covalently linked to the other protein at the N-terminus of the constant domain.
  • such fusion proteins may facilitate purification, increase half -life in vivo, and enhance the delivery of an antigen across an epithelial barrier to the immune system.
  • the immunoglobulins of the invention include analogs and derivatives that are either modified, i.e, by the covalent attachment of any type of molecule as long as such covalent attachment that does not impair immunospecific binding.
  • the derivatives and analogs of the immunoglobulins include those that have been further modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, etc. Additionally, the analog or derivative may contain one or more non-classical amino acids.
  • the foregoing antibodies can be used in methods known in the art relating to the localization and activity of the MSPIs of the invention, e.g., for imaging these polypeptides, measuring levels thereof in appropriate physiological samples, in diagnostic methods, etc. 5.12 EXPRESSION OF ANTIBODIES
  • the antibodies of the invention can be produced by any method l ⁇ iown in the art for the synthesis of antibodies, in particular, by chemical synthesis or by recombinant expression, and are preferably produced by recombinant expression techniques. Recombinant expression of antibodies, or fragments, derivatives or analogs thereof, requires construction of a nucleic acid that encodes the antibody.
  • a nucleic acid encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., 1994, BioTechniques 17:242), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding antibody, annealing and ligation of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
  • chemically synthesized oligonucleotides e.g., as described in Kutmeier et al., 1994, BioTechniques 17:242
  • oligonucleotides e.g., as described in Kutmeier et al., 1994, BioTechniques 17:242
  • the nucleic acid encoding the antibody may be obtained by cloning the antibody. If a clone containing the nucleic acid encoding the particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic, acid encoding the antibody may be obtained from a suitable source (e.g., an antibody cDNA library, or cDNA library generated from any tissue or cells expressing the antibody) by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence.
  • a suitable source e.g., an antibody cDNA library, or cDNA library generated from any tissue or cells expressing the antibody
  • antibodies specific for a particular antigen may be generated by any method known in the art, for example, by immunizing an animal, such as a rabbit, to generate polyclonal antibodies or, more preferably, by generating mAbs.
  • a clone encoding at least the Fab portion of the antibody may be obtained by screening Fab expression libraries (e.g., as described in Huse et al., 1989, Science 246:1275-1281) for clones of Fab fragments that bind the specific antigen or by screening antibody libraries (See, e.g., Clackson et al., 1991, Nature 352:624; Hane et al., 1997 Proc. Natl. Acad. Sci. USA 94:4937).
  • nucleic acid encoding at least the variable domain of the antibody molecule may be introduced into a vector containing the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., WO 86/05807; WO 89/01036; and U.S. 5,122,464).
  • Vectors containing the complete light or heavy chain for co-expression with the nucleic acid to allow the expression of a complete antibody molecule are also available.
  • the nucleic acid encoding the antibody can be used to introduce the nucleotide substitution(s) or deletion(s) necessary to substitute (or delete) the one or more variable region cysteine residues participating in an intrachain disulfide bond with an amino acid residue that does not contain a sulfhydyl group.
  • Such modifications can be carried out by any method known in the art for the introduction of specific mutations or deletions in a nucleotide sequence, for example, but not limited to, chemical mutagenesis, in vitro site directed mutagenesis (Hutchinson et al., 1978, J. Biol. Chem. 253:6551), PCR based methods, etc.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human antibody constant region, e.g., humanized antibodies.
  • the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art.
  • methods for preparing the protein of the invention by expressing nucleic acid containing the antibody molecule sequences are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing an antibody molecule coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. See, for example, the techniques described in Sambrook et al.
  • the expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody of the invention.
  • the host cells used to express a recombinant antibody of the invention may be either bacterial cells such as Escherichia coli, or, preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., 198, Gene 45:101; Cockett et al., 1990, Bio/Technology 8:2).
  • host-expression vector systems may be utilized to express an antibody molecule of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express the antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid).
  • yeast e.g., Saccharomyces, Pichia
  • insect cell systems infected with recombinant virus expression vectors e.g., baculovirus
  • plant cell systems infected with recombinant virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus
  • mammalian cell systems e.g., COS, CHO, BHK, HEK293, 3T3 cells harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • mammalian cells e.g., COS, CHO, BHK, HEK293, 3T3 cells
  • promoters derived from the genome of mammalian cells e.g., metallothionein promoter
  • mammalian viruses e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter.
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J. 2: 1791), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pJN vectors (Inouye & Inouye, 1985, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutafhione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety. . .
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is. used as a vector to express foreign genes.
  • the virus grows in Spodopterafrugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • an AcNPV promoter for example the polyhedrin promoter.
  • a number of viral-based expression systems e.g., an adenovirus expression system
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • cell lines that stably express an antibody of interest can be produced by transfecting the cells with an expression vector comprising the nucleotide sequence of the antibody and the nucleotide sequence of a selectable marker (e.g., neomycin or hygromycin), and selecting for expression of the selectable marker.
  • a selectable marker e.g., neomycin or hygromycin
  • the expression levels of the antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing antibody is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., 1983, Mol. Cell. Biol. 3:257).
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; Kohler, 1980, Proc. Natl. Acad. Sci. USA 77:2197).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA. .
  • the antibody molecule of the invention may be purified by any method known in the art for purification of an antibody molecule, for example, by chromatography (e.g., ion exchange chromatography, affinity chromatography such as with protein A or specific antigen, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • any fusion protein may be readily purified by utilizing an antibody specific for the fusion protein being expressed. For example, a system described by Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad.
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues.
  • the tag serves as a matrix binding domain for the fusion protein. Extracts from cells infected- with recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose columns and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • anti-MSPI antibodies or fragments thereof are conjugated to a diagnostic or therapeutic moiety.
  • the antibodies can be used for diagnosis or to determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive nuclides, positron emitting metals (for use in positron emission tomography), and nonradioactive paramagnetic metal ions. See generally U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • Suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; suitable prosthetic groups include • streptavidin, avidin and biotin; suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride and phycoerythrin; suitable luminescent materials include luminol; suitable bioluminescent materials include luciferase, luciferin, and aequorin; and suitable radioactive nuclides include 1251, 1311, 11 lln and 99Tc.
  • an anti-MSPI antibodies or fragments thereof can be conjugated to a therapeutic agent or drug moiety to modify a given biological response.
  • the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents, e.g., small molecules.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha -interferon, beta -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, a biological .
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, alpha -interferon, beta -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin
  • angiostatin or endostatin e
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • NGF nerve growth factor
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described in U.S. 4,676,980.
  • An antibody with or without a therapeutic moiety conjugated to it can be used as a therapeutic that is administered alone or in combination with cytokine(s).
  • a first sample of CSF, serum, plasma or urine obtained from a subject suspected of having or known to have MS can be used for diagnosis or monitoring.
  • a decreased abundance of one or more MSFs or MSPIs (or any combination of them) in a first sample or sample set relative to a second sample or sample set (from a subject or subjects free from MS) or a previously determined reference range indicates the presence of MS ;
  • MSFs and MSPIs suitable for this purpose are identified in Tables I and IV, respectively, as described in detail above.
  • an increased abundance of one or more MSFs or MSPIs (or any combination of them) in a first sample or sample set compared to a second sample or sample set or a previously determined reference range indicates the presence of MS; MSFs and MSPIs suitable for this purpose are identified in Tables ⁇ and V, respectively, as described in detail above.
  • the relative abundance of one or more MSFs or MSPIs (or any combination of them) in a first sample or sample set compared to a second sample or sample set or a previously determined reference range indicates a subtype of MS (e.g., benign or progressive MS).
  • the relative abundance of one or more MSFs or MSPIs (or any combination of them) in a first sample or sample set relative to a second sample or sample set or a previously determined reference range indicates the degree or severity of MS.
  • detection of one or more MSPIs described herein may optionally be combined with detection of one or more additional biomarkers for MS including, but not limited to, oligoclonal immunoglobulin bands in CSF revealed by isoelectric focusing (Reiber H et al. (1998) Mult Scler 3: 111-7).
  • any suitable method in the art can be employed to measure the level of MSFs and MSPIs, including but not limited to the Preferred Technology described herein, kinase assays, irnmunoassays to detect and/or visualize the MSPIs (e.g., Western blot, immunoprecipitation followed by sodium dodecyl sulfate polyacrylamide gel electrophoresis, immunocytochemistry, etc.).
  • an assay for that function may be used to measure MSPI expression.
  • a decreased abundance of mRNA including one or more MSPIs identified in Table IV (or any combination of them) in a first sample or sample set relative to a second sample or sample set or a previously determined reference range indicates the presence of MS.
  • an increased abundance of mRNA encoding one or more MSPIs identified in Table V (or any combination of them) in a first sample or sample set relative to a second sample or sample set or previously determined reference range indicates the presence of MS.
  • Any suitable hybridization assay can be used to detect MSPI expression by detecting and or visualizing mRNA encoding the MSPI (e.g., Northern assays, dot blots, in situ hybridization, etc.).
  • labeled antibodies, derivatives and analogs thereof, which specifically bind to an MSPI can be used for diagnostic purposes to detect, diagnose, or monitor MS.
  • MS is detected in an animal, more preferably in a mammal and most preferably in a human.
  • the invention provides methods for identifying agents (e.g., candidate agents) that bind to an MSPI or have a stimulatory or inhibitory effect on the expression or activity of an MSPI.
  • the invention also provides methods of identifying agents or candidate agents that bind to an MSPI fragment, MSPI-related polypeptide or the MSPI-fusion protein or have a stimulatory or inhibitory effect on the expression or activity of an MSPI fragment, MSPI-related polypeptide or an MSPI fusion protein.
  • agents or candidate agents include, but are not limited to, nucleic acids (e.g., DNA and RNA), carbohydrates, lipids, proteins, peptides, peptidomimetics, small molecules and other drugs.
  • Agents can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection.
  • biological libraries include biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, 1997, Anticancer Drug Des. 12: 145; U.S. 5,738,996; and U.S. 5,807,683).
  • Libraries of compounds may be presented, e.g., presented in solution (e.g., Houghten, 1992, Bio/Techniques 13:412-421), or on beads (Lam, 1991, Nature 354:82-84), chips (Fodor, 1993, Nature 364:555-556), bacteria (U.S. Patent No. 5,223,409), spores (US Patent Nos.
  • agents that interact with (i.e., bind to) an MSPI, an MSPI fragment e.g.
  • an MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein are identified in a cell-based assay system.
  • cells expressing an MSPI, a fragment of an MSPI, an MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein are contacted with a candidate agent or a control agent and the ability of the candidate agent to interact with the MSPI is determined.
  • this assay may be used to screen a plurality (e.g. a library) of candidate agents.
  • the cell for example, can be of prokaryotic origin (e.g., E.
  • the cells can express the MSPI, fragment of the MSPI, MSPI-related polypeptide, a fragment of the MSPI-related polypeptide, or an MSPI fusion protein endogenously or be genetically engineered to express the MSPI, fragment of the MSPI, MSPI-related polypeptide, a fragment of the MSPI-related polypeptide, or an MSPI fusion protein.
  • the MSPI, fragment of the MSPI, MSPI-related polypeptide, a fragment of the MSPI-related polypeptide, or an MSPI fusion protein or the candidate agent is labeled, for example with a radioactive label (such as 32P, 35S or 1251) or a fluorescent label (such as fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde or fluorescamine) to enable detection of an interaction between an MSPI and a candidate agent.
  • a radioactive label such as 32P, 35S or 1251
  • a fluorescent label such as fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde or fluorescamine
  • the ability of the candidate agent to interact directly or indirectly with an MSPI, a fragment of an MSPI, an MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein can be determined by methods known to those of skill in the art.
  • the interaction between a candidate agent and an MSPI, a fragment of an MSPI, an MSPI-related- polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein can be determined by flow cytometry, a scintillation assay, immunoprecipitation or western blot analysis.
  • agents that interact with (i.e., bind to) an MSPI, an MSPI fragment (e.g., a functionally active fragment) an MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein are identified in a cell-free assay system.
  • a native or recombinant MSPI or fragment thereof, or a native or recombinant MSPI-related polypeptide or fragment thereof, or an MSPI-fusion protein or fragment thereof is contacted with a candidate agent or a control agent and the ability of the candidate agent to interact with the MSPI or MSPI-related polypeptide, or MSPI fusion protein is determined.
  • this assay may be used to screen a plurality (e.g. a library) of candidate agents.
  • the MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI-fusion protein is first immobilized, by, for example, contacting the MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein with an immobilized antibody which specifically recognizes and binds it, or by contacting a purified preparation of the.
  • the MSPI, MSPI fragment, MSPI- related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein may be partially or completely purified (e.g., partially or completely free of other polypeptides) or part of a cell lysate. Further, the MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide may be a fusion protein comprising the MSPI or a biologically active portion thereof, or MSPI-related polypeptide and a domain such as glutathionine-S-transferase.
  • MSPI, MSPI fragment, MSPI-related polypeptide, fragment of an MSPI-related polypeptide or MSPI fusion protein can be biotinylated using techniques well known to those of skill in the art (e.g., biotinylation kit, Pierce Chemicals; Rockford, JL).
  • biotinylation kit Pierce Chemicals; Rockford, JL.
  • the ability of the candidate agent to interact with an MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or. an . MSPI fusion protein can be determined by methods known to those of skill in the art.
  • a cell-based assay system is used to identify agents that bind to or modulate the activity of a protein, such as an enzyme, or a biologically active portion thereof, which is responsible for the production or degradation of an MSPI or is responsible for the post- translational modification of an MSPI.
  • a protein such as an enzyme, or a biologically active portion thereof, which is responsible for the production or degradation of an MSPI or is responsible for the post- translational modification of an MSPI.
  • a plurality e.g., a library
  • candidate agents are contacted with cells that naturally or recombinantly express: (i) an MSPI, an MSPI homolog an MSPI-related polypeptide, an MSPI fusion protein, or a biologically active fragment of any of the foregoing; and (ii) a protein that is responsible for processing of the MSPI, MSPI homolog, MSPI-related polypeptide, MSPI fusion protein, or fragment in order to identify agents that modulate the production, degradation, or post-translational modification of the MSPI, MSPI homolog, MSPI-related polypeptide, MSPI ' fusion protein or fragment.
  • candidate agents identified in the primary screen can then be assayed in a secondary screen against cells naturally or recombinantly expressing the specific MSPIs of interest.
  • the ability of the candidate agent to modulate the production, degradation or post-translational modification of an MSPI, homolog, MSPI-related polypeptide, or MSPI fusion protein can be determined by methods known to those of skill in the art, including without limitation, flow cytometry, a scintillation assay, immunoprecipitation and western blot analysis.
  • agents that competitively interact with (i.e., bind to) an MSPI bind to
  • MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein are identified in a competitive binding assay.
  • cells expressing an MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein are contacted with a candidate agent and a agent known to interact with the MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide or an MSPI fusion protein; the ability of the candidate agent to competitively interact with the MSPI, MSPI fragment, MSPI-related polypeptide, fragment of an MSPI-related polypeptide, or an MSPI fusion protein is then determined.
  • agents that competitively interact with (i.e., bind to) an MSPI, MSPI fragment, MSPI-related polypeptide or fragment of an MSPI-related polypeptide are identified in a cell-free assay system by contacting an MSPI, MSPI fragment, MSPI-related polypeptide, fragment of an MSPI-related polypeptide, or an MSPI fusion protein with a candidate agent and a agent known to interact with the MSPI, MSPI-related polypeptide or MSPI fusion protein.
  • the ability of the candidate agent to interact with an MSPI, MSPI fragment, MSPI-related polypeptide, a fragment of an MSPI-related polypeptide, or an MSPI fusion protein can be determined by methods known to those of skill in the art.
  • These assays, whether cell-based or cell-free can be used to screen a plurality (e.g., a library) of candidate agents.
  • agents that modulate i.e., upregulate or downregulate the expression of an MSPI, or an MSPI-related polypeptide are identified by contacting cells (e.g., cells of prokaryotic origin or eukaryotic origin) expressing the MSPI, or MSPI-related polypeptide with a candidate agentor a control agent (e.g., phosphate buffered saline (PBS)) and determining the expression of the MSPI, MSPI-related polypeptide, or MSPI fusion .
  • a candidate agentor a control agent e.g., phosphate buffered saline (PBS)
  • the level of expression of a selected MSPI, MSPI-related polypeptide, mRNA encoding the MSPI, or mRNA encoding the MSPI-related polypeptide in the presence of the candidate agent is compared to the level of expression of the MSPI, MSPI-related polypeptide, mRNA encoding the MSPI, or mRNA encoding the MSPI-related polypeptide in the absence of the candidate agent (e.g., in the presence of a control agent).
  • the candidate agent can then be identified as a modulator of the expression of the MSPI, or an MSPI-related polypeptide based on this comparison.
  • the candidate agent when expression of the MSPI or mRNA is significantly greater in the presence of the candidate agent than in its absence, the candidate agent is identified as a stimulator of expression of the MSPI or mRNA.
  • the candidate agent when expression of the MSPI or mRNA is significantly less in the presence of the candidate agent than in its absence, the candidate agent is identified as an inhibitor of the expression of the MSPI or mRNA.
  • the level of expression of an MSPI or the mRNA that encodes it can be determined by methods known to those of skill in the art. For example, mRNA expression can be assessed by Northern blot analysis or RT-PCR, and protein levels can be assessed by western blot analysis.
  • the candidate agent may be an agonist or an antagonist of the MSPI, MSPI-related polypeptide, or MSPI fusion protein, or of an upstream effector of the MSPI, MSPI-related polypeptide or MSPI fusion protein.
  • agents that modulate the activity of an MSPI, or an MSPI-related polypeptide are identified by contacting a preparation containing the MSPI or MSPI-related polypeptide, or cells (e.g., prokaryotic or eukaryotic cells) expressing the MSPI or MSPI-related polypeptide with a candidate agent or a control agent and determining the ability of candidate agent to modulate (e.g., stimulate or inhibit) the activity of the MSPI or MSPI-related polypeptide.
  • an MSPI or an MSPI-related polypeptide can be assessed by detecting changes in a downstream effector such as, without limitation, induction of a cellular signal transduction pathway of the MSPI or MSPI-related polypeptide (e.g., .intracellular Ca2 + , diacylglycerol, JP3, etc.), detecting catalytic or enzymatic activity of the target on a suitable substrate, detecting the induction of a reporter gene (e.g., a regulatory, element that is responsive to an MSPI or an MSPI-related polypeptide and is operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cellular differentiation, or cell proliferation.
  • a reporter gene e.g., a regulatory, element that is responsive to an MSPI or an MSPI-related polypeptide and is operably linked to a nucleic acid encoding a detectable marker, e.g.
  • the candidate agent can then be identified as a modulator of the activity, of an MSPI or MSPI-related . polypeptide by comparing the effects of the candidate agent to the control agent.
  • Suitable control agents include phosphate buffered saline (PBS) and normal saline (NS).
  • agents that modulate i.e., upregulate or downregulate) the . expression, activity or both the expression and activity of an MSPI or MSPI-related polypeptide are identified in an animal model.
  • suitable animals include, but are not limited to, mice, rats, rabbits, monkeys, guinea pigs, dogs and cats.
  • the animal used represent a model of MS (e.g., experimental autoimmune encephalomyelitis (EAE) (Steinman (1999) Neuron, 24:511-514)).
  • EAE experimental autoimmune encephalomyelitis
  • the candidate agent or a control agent is administered (e.g., orally, rectally or parenterally such as intraperitoneally or intravenously) to a suitable animal and the effect on the expression, activity or both expression and activity of the MSPI or MSPI-related polypeptide is determined. Changes in the expression of an MSPI or MSPI-related polypeptide can be assessed by the methods outlined above.
  • an MSPI or MSPI-related polypeptide is used as a "bait protein" in a two-hybrid assay or three hybrid assay to identify other proteins that bind to or interact with an MSPI or MSPI-related polypeptide (see, e.g., U.Si Patent No. 5,283,317;
  • binding proteins may be involved in the propagation of signals by the MSPIs of the invention as, for example, upstream or downstream elements of a signaling pathway involving the MSPIs of the invention.
  • Table IX enumerates scientific publications describing suitable assays for detecting or quantifying enzymatic or binding activity of an MSPI, an MSPI analog, an MSPI-related polypeptide, or a fragment of any of the foregoing. Each such reference is hereby incorporated in its entirety.
  • as assay referenced in Table IX is used in the screens and assays described herein, for example to screen for or identify candidate agent that modulates the activity and or expression of an MSPI, MSPI analog, or MSPI-related polypeptide, a fragment of any of the foregoing.
  • This invention further provides novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
  • the invention provides for treatment or prevention of various diseases and disorders by administration of a therapeutic agent.
  • agents include but are not limited to: MSPIs, MSPI analogs, MSPI-related polypeptides and derivatives (including fragments) thereof; antibodies to the foregoing; nucleic acids encoding MSPIs, MSPI analogs, MSPI-related polypeptides and fragments thereof; antisense nucleic acids to a gene encoding an MSPI or MSPI-related polypeptide; and modulator (e.g., agonists and antagonists) of a gene encoding an MSPI or MSPI-related polypeptide.
  • modulator e.g., agonists and antagonists
  • An important feature of the present invention is the identification of genes encoding MSPIs involved in MS. MS can be treated (e.g.
  • one or more antibodies each specifically binding to an MSPI are administered alone or in combination with one or more additional therapeutic agents or treatments.
  • Such therapeutic agents or treatments include, but are not limited to, Interferon ⁇ -lb (BetaseronTM, BetaferonTM), Interferon ⁇ -la (AvonexTM, RebifTM), Glatiramer acetate (CopaxoneTM), intravenous immunoglobulin and, for incidences of acute relapse, therapies with corticosteroids (Noseworthy (1999) Nature 399: suppl. A40-A47).
  • a biological product such as an antibody is allogeneic to the subject to which it is administered.
  • a human MSPI or a human MSPI-related polypeptide, a nucleotide sequence encoding a human MSPI or a human MSPI-related polypeptide, or an antibody to a human MSPI or a human MSPI-related polypeptide is administered to a human subject for therapy (e.g. to ameliorate symptoms or to retard onset or progression) or prophylaxis.
  • MS is treated or prevented by administration to a subject suspected of having or l nown to have MS or to be at risk of developing MS of a agent that modulates (i.e., increases or decreases) the level or activity (i.e., function) of one or more MSPIs ⁇ or the level of one or more MSFs - that are differentially present in the CSF of subjects having MS compared with CSF of subjects free from MS.
  • a agent that modulates (i.e., increases or decreases) the level or activity (i.e., function) of one or more MSPIs ⁇ or the level of one or more MSFs - that are differentially present in the CSF of subjects having MS compared with CSF of subjects free from MS.
  • MS is treated or prevented by administering to a subject suspected of having or known to have MS or to be at risk of developing MS a agent that upregulates (i.e., increases) the level or activity (i.e., function) of one or more MSPIs - or the level of one or more MSFs - that are decreased in the CSF of subjects having MS.
  • a agent is administered that upregulates the level or activity (i.e., function) of one or more MSPIs - or the level of one or more MSFs - that are increased in the CSF of subjects having MS.
  • Such a agent examples include but are not limited to: MSPIs, MSPI fragments and MSPI-related polypeptides; nucleic acids encoding an MSPI, an MSPI fragment and an MSPI-related polypeptide (e.g., for use in gene therapy); and, for those MSPIs or MSPI-related polypeptides with enzymatic activity, agents or molecules known to modulate that enzymatic activity.
  • Other agents that can be used, e.g., MSPI agonists can be identified using in vitro assays.
  • MS is also treated or prevented by administration to a subject suspected of having or known to have MS or to be at risk of developing MS of a agent that downregulates the level or activity of one or more MSPIs - or the level of one or more MSFs - that are increased in the CSF of subjects having MS.
  • a agent is administered that downregulates the level or activity of one or more MSPIs - or the level of one or more MSFs - that are decreased in the CSF of subjects having MS.
  • examples of such a agent include, but are not limited to, MSPI antisense oligonucleotides, ribozymes, antibodies directed against MSPIs, and agents that inhibit the enzymatic activity of an MSPI.
  • Other useful agents e.g., MSPI antagonists and small molecule MSPI antagonists, can be identified using in vitro assays.
  • agents that promote the level or function of one or more MSPIs, or the level of one or more MSFs are therapeutically or prophylactically administered to a subject suspected of having or known to have MS, in whom the levels or functions of said one or more MSPIs, or levels of said one or more MSFs, are absent or are decreased relative to a control or normal reference range.
  • agents that promote the level or function of one or more MSPIs, or the level of one or more MSFs are therapeutically or prophylactically administered to a subject suspected of having or known to have MS in whom the levels or functions of said one or more MSPIs, or levels of said one or more MSFs, are increased relative to a control or to a reference range.
  • agents that decrease the level or function of one or more MSPIs, or the level of one or more MSFs are therapeutically or prophylactically administered to a subject suspected of having or known to have MS in whom the levels or functions of said one or more MSPIs, or levels of said one or more MSFs, are increased relative to a control or to a reference range.
  • agents that decrease the level or function of one or more MSPIs, or the level of one or more MSFs are therapeutically or prophylactically administered to a subject suspected of having or known to have MS in whom the levels or functions of said one or more MSPIs, or levels of said one or more MSFs, are decreased relative to a control or to a reference range.
  • the change in MSPI function or level, or MSF level, due to the administration of such agents can be readily detected, e.g., by obtaining a sample (e.g., a sample of CSF, blood or urine or a tissue sample such as biopsy tissue) and assaying in vitro the levels of said MSFs or the levels or activities of said MSPIs, or the levels of mRNAs encoding said MSPIs. or any combination of the foregoing. Such assays can be performed before and after the administration of the agent as described herein.
  • the agents of the invention include but are not limited to any agent, e.g., a small organic molecule, protein, peptide, antibody, nucleic acid, etc.
  • Interferon ⁇ -lb BetaseronTM, BetaferonTM
  • Interferon ⁇ -la AvonexTM, RebifTM
  • Glatiramer acetate CopaxoneTM
  • intravenous immunoglobulin and, for incidences of acute relapse, therapies with corticosteroids (Noseworthy (1999) Nature 399:suppl. A40-A47).
  • nucleic acids comprising a sequence encoding an MSPI, an MSPI fragment, MSPI-related polypeptide or fragment of an MSPI-related polypeptide, are administered to promote MSPI function by way of gene therapy.
  • Gene therapy refers to administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acid produces its encoded polypeptide that mediates a therapeutic effect by promoting MSPI function.
  • the agent comprises a nucleic acid encoding an MSPI or fragment or chimeric protein thereof, said nucleic acid being part of an expression vector that expresses an MSPI or fragment or chimeric protein thereof in a suitable host.
  • such a nucleic acid has a promoter operably linked to the MSPI coding region, said promoter being inducible or constitutive (and, optionally, tissue-specific).
  • a nucleic acid molecule is used in which the MSPI coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a ⁇ desired site in the genome, thus providing for intrachromosomal expression of the MSPI nucleic acid (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438).
  • Delivery of the nucleic acid into a subject may be direct, in which case the subject is directly exposed to the nucleic acid or nucleic acid-carrying vector; this approach is known as in vivo gene therapy.
  • delivery of the nucleic acid into the subject may be indirect, in which case cells are first transformed with the nucleic acid in vitro and then transplanted into the subject; this approach is known as ex vivo gene therapy.
  • the nucleic acid is directly administered in vivo, where it is expressed to produce the encoded product.
  • BiolisticTM, Dupont by coating with lipids, cell-surface receptors or transfecting agents; by encapsulation in liposomes, microparticles or microcapsules; by administering it in linkage to a peptide which is known to enter the nucleus; or by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432),' which can be used to target cell types specifically expressing the receptors.
  • a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., WO 92/06180, WO 92/22635, WO92/20316, W093/14188, WO 93/20221).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438).
  • a viral vector that contains a nucleic acid encoding an MSPI is used.
  • a retroviral vector can be used (see Miller et al., 1993, Meth. Enzymol. 217:581-599). These retroviral vectors have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
  • the nucleic acid encoding the MSPI to be used in gene therapy is cloned into the vector, which facilitates delivery of the gene into a subject.
  • retroviral vectors More detail about retroviral vectors can be found in Boesen et al., 1994, Biotherapy 6:291-302, which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy.
  • Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., 1994, J. Clin. Invest. 93:644-651; Kiem et al., 1994, Blood 83:1467-1473; Salmons and Gunzberg, 1993, Human Gene Therapy 4:129-141; and Grossman and Wilson, 1993, Curr. Opin. in Genetics and Devel. 3:110-114.
  • Adenoviruses are other viral vectors that can be used in gene therapy. Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenoviruses • naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, 1993, Current Opinion in Genetics and Development 3:499-503. present a review of adenovirus-based gene therapy.
  • Adeno-associated virus has also been proposed for use in gene therapy (Walsh et al, 1993, Proc. Soc. Exp. Biol. Med. 204:289-300; U.S. Patent No. 5,436,146).
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a subject.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc. Numerous techniques are known in the art for the.introduction of foreign genes into cells (see, e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618; Cohen et al., 1993, Meth. Enzymol.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a subject by various methods known in the art.
  • epithelial cells are injected, e.g., subcutaneously.
  • recombinant skin cells may be applied as a skin graft onto the subject.
  • Recombinant blood cells e.g., hematopoietic stem or progenitor cells
  • the amount of cells envisioned for use depends on the desired effect, the condition of the subject, etc., and can be determined by one skilled in the art.
  • Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to neuronal cells, glial cells (e.g., oligodendrocytes or astrocytes), epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood or fetal liver.
  • the cell used for gene therapy is autologous to the subject that is treated.
  • a nucleic acid encoding an MSPI is introduced into the cells such that it is expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem or progenitor cells which can be isolated and maintained in vitro can be used in accordance with this embodiment of the present invention (see e.g. WO 94/08598; Stemple and Anderson, 1992, Cell 71:973-985; Rheinwald, 1980, Meth. Cell Bio. 21A:229; and Pittelkow and Scott, 1986, Mayo Clinic Proc. 61:771).
  • the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • Direct injection of a DNA coding for an MSPI may also be performed according to, for example, the techniques described in United States Patent No. 5,589,466. These techniques involve the injection of "naked DNA", i.e., isolated DNA molecules in the absence of liposomes, cells, or any other material besides a suitable carrier.
  • naked DNA i.e., isolated DNA molecules in the absence of liposomes, cells, or any other material besides a suitable carrier.
  • the injection of DNA encoding a protein and operably linked to a suitable promoter results in the production of the protein in cells near the site of injection and the elicitation of an immune response in the subject to the protein encoded by the injected DNA.
  • naked DNA comprising (a) DNA encoding an MSPI and (b) a promoter are injected into a subject to elicit an immune response to the MSPI.
  • MS is treated or prevented by administration of a agent that antagonizes (inhibits) the level(s) and/or function(s) of one or more MSPIs which are elevated in a body fluid sample of subjects having MS as compared with a body fluid sample of subjects free from MS.
  • Agents useful for this purpose include but are not limited to anti-MSPI antibodies (and fragments and derivatives containing the binding region thereof), MSPI antisense or ribozyme nucleic acids, and nucleic acids encoding dysfunctional MSPIs that are used to "J ⁇ iockout" endogenous MSPI function by homologous recombination (see, e.g., Capecchi, 1989, Science 244:1288-1292).
  • agents that inhibit MSPI function can be identified by use of known in vitro assays, e.g., assays for the ability of a test agent to inhibit binding of an MSPI to another protein or a binding partner, or to inhibit a known MSPI function. Preferably such inhibition is assayed in vitro or in cell culture, but genetic assays may also be employed.
  • the Preferred Technology can also be used to detect levels of the MSPIs before and after the administration of the agent.
  • suitable in vitro or in vivo assays are utilized to determine the effect of a specific agent and whether its administration is indicated for treatment of the affected tissue, as described in more detail below.
  • a agent that inhibits an MSPI function is administered therapeutically or prophylactically to a subject in whom an increased CSF level or functional activity of the MSPI (e.g., greater than the normal level or desired level) is detected as compared with CSF of subjects free from MS or a predetermined reference range.
  • an increased CSF level or functional activity of the MSPI e.g., greater than the normal level or desired level
  • Methods standard in the art can be employed to measure the increase in an MSPI level or function, as outlined above.
  • Preferred MSPI inhibitor compositions include small molecules, i.e., molecules of 1000 daltons or less. Such small molecules can be identified by the screening methods described herein.
  • an MSPI may be seen to be decreased in the CSF where this decrease represents an increase in the MSPI level in another compartment, for example but without limitation sequestering of the MSPI in a cell, subcellular compartment, body fluid or tissue rather than secretion into CSF.
  • a agent that inhibits an MSPI function is administered therapeutically or prophylactically to a subject in whom a decreased CSF level or functional activity of the MSPI (e.g., greater than the normal level or desired level) is detected as compared with CSF of subjects free from MS or a predetermined reference range.
  • MSPI expression is inhibited by use of MSPI antisense nucleic acids.
  • the present invention provides the therapeutic or prophylactic use of nucleic acids comprising at least six nucleotides that are antisense to a gene or cDNA encoding an MSPI or a portion thereof.
  • an MSPI "antisense" nucleic acid refers to a nucleic acid capable of hybridizing by virtue of some sequence complementarity to a portion of an RNA (preferably mRNA) encoding an MSPI.
  • the antisense nucleic acid may be complementary to a coding and/or noncoding region of an mRNA encoding an MSPI.
  • antisense nucleic acids have utility as agents that inhibit MSPI expression, and can be used in the treatment or prevention of MS.
  • the antisense nucleic acids of the invention are double-stranded or single-stranded oligonucleotides, RNA or DNA or a modification or derivative thereof, and can be directly administered to a cell or produced intracellularly by transcription of exogenous, introduced sequences.
  • the invention further provides pharmaceutical compositions comprising an effective amount of the MSPI antisense nucleic acids of the invention in a pharmaceutically acceptable carrier, as described infra.
  • the invention provides methods for inhibiting the expression of an MSPI nucleic acid sequence in a prokaryotic or eukaryotic cell comprising providing the cell with an effective amount of a composition comprising an MSPI antisense nucleic acid of the invention.
  • MSPI antisense nucleic acids and their uses are described in detail below.
  • the MSPI antisense nucleic acids are of at least six nucleotides and are preferably oligonucleotides ranging from 6 to about 50 oligonucleotides.
  • the oligonucleotide is at least 10 nucleotides, at least 15 nucleotides, at least 100 nucleotides, or at least 200 nucleotides.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof and can be single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide may include other appended groups such as peptides; agents that facilitate transport across the cell membrane (see, e.g., Letsinger et al., 1989, Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al., 1987, Proc. Natl. Acad. Sci.
  • an MSPI antisense oligonucleotide is provided, preferably of single-stranded DNA.
  • the oligonucleotide may be modified at any position on . its structure with substituents generally known in the art.
  • the MSPI antisense oligonucleotide may comprise at least one of the following modified base moieties: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmefhyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueo
  • the oligonucleotide comprises at least one modified sugar moiety, e.g., one of the following sugar moieties: arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the oligonucleotide comprises at least one of the following modified phosphate backbones: a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, a formacetal, or an analog of formacetal.
  • the oligonucleotide is an alpha-anomeric oligonucleotide.
  • An alpha-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual alpha -units, the strands run parallel to each other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641).
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent.
  • Oligonucleotides of the invention may be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.).
  • an automated DNA synthesizer such as are commercially available from Biosearch, Applied Biosystems, etc.
  • phosphorothioate oligonucleotides may be synthesized by the method of Stein et al. (1988, Nucl. Acids Res. 16:3209), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., 1988, Proc. Natl. Acad. Sci. USA 85:7448-7451).
  • the MSPI antisense nucleic acid of the invention is produced intracellularly by transcription from an exogenous sequence.
  • a vector can be introduced in vivo such that it is taken up by a cell, within which cell the vector or a portion thereof is transcribed, producing an antisense nucleic acid (RNA) of the invention.
  • RNA antisense nucleic acid
  • Such a vector would contain a sequence encoding the MSPI antisense nucleic acid.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA technology standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the MSPI antisense RNA can be by any promoter known in the art to act in mammalian, preferably human, cells. Such promoters can be inducible or constitutive. Examples of such promoters are outlined above.
  • the antisense nucleic acids of the invention comprise a sequence complementary to at least a portion of an RNA transcript of a gene encoding an MSPI, preferably a human gene encoding an MSPI. However, absolute complementarity, although preferred, is not required.
  • a sequence "complementary to at least a portion of an RNA,” as referred to herein, means a sequence having sufficient complementarity to be able to hybridize under stringent conditions (e.g., highly stringent conditions or moderately stringent conditions as defined supra) with the RNA, forming a stable duplex; in the case of double-stranded MSPI antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • stringent conditions e.g., highly stringent conditions or moderately stringent conditions as defined supra
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid.
  • the longer the hybridizing nucleic acid the more base mismatches with an RNA encoding an MSPI it may contain and still form a stable duplex (or triplex, as the case may be).
  • One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
  • MSPI antisense nucleic acids can be used to treat or prevent MS when the target
  • MSPI is overexpressed in the CSF of subjects suspected of having or suffering from MS.
  • a single-stranded DNA antisense MSPI oligonucleotide is used.
  • Cell types which express or oyerexpress RNA encoding an MSPI can be identified by various methods known in the art. Such cell types include but are not limited to leukocytes (e.g., neutrophils, macrophages, monocytes) and resident cells (e.g., astrocytes, glial cells, neuronal cells, and ependymal cells). Such methods include, but are not limited to, hybridization with an MSPI-specific nucleic acid (e.g., by Northern hybridization, dot blot hybridization, in situ hybridization), observing the ability of RNA from the cell type to be translated in vitro into an MSPI, immunoassay, etc. In a preferred aspect, primary tissue from a subject can be assayed for MSPI expression prior to treatment, e.g., by immunocytochemistry or in situ hybridization.
  • leukocytes e.g., neutrophils, macrophages, monocytes
  • resident cells e.g., astrocytes, glial cells
  • compositions of the invention comprising an effective amount of an MSPI antisense nucleic acid in a pharmaceutically acceptable carrier, can be administered to a subject having MS.
  • MSPI antisense nucleic acid The amount of MSPI antisense nucleic acid which will be effective in the treatment of MS can be determined by standard clinical techniques.
  • compositions comprising one or more MSPI antisense nucleic acids are administered via liposomes, microparticles, or microcapsules.
  • such compositions may be used to achieve sustained release of the MSPI antisense nucleic acids.
  • symptoms of MS may be ameliorated by decreasing the level of an MSPI or MSPI activity by using gene sequences encoding the MSPI in conjunction with •well-known gene "knock-out,” ribozyme or triple helix methods to decrease gene expression of an MSPI.
  • ribozyme or triple helix molecules are used to modulate the activity, expression or synthesis of the gene encoding the MSPI, and thus to ameliorate the symptoms of MS.
  • Such molecules may be designed to reduce or inhibit expression of a mutant or non-mutant target gene. Techniques for the production and use of such molecules are well known to those of skill in the art.
  • Ribozyme molecules designed to catalytically cleave gene mRNA transcripts encoding an MSPI can be used to prevent translation of target gene mRNA and, therefore, expression of the gene product.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. (For a review, see Rossi, 1994, Current Biology 4, 469-471). The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage event.
  • composition of ribozyme molecules must include one or more sequences complementary to the target gene mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see, e.g., U.S. Patent No. 5,093,246, which is inco ⁇ orated herein by reference in its entirety.
  • ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy mRNAs encoding an MSPI
  • the use of hammerhead ribozymes is preferred.
  • Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA have the following sequence of two bases: 5'-UG-3'.
  • the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the mRNA encoding the MSPI, i.e., to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.
  • the ribozymes of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes”) such as the one that occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and that has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224, 574-578; Zaug and Cech, 1986, Science, 231, 470-475; Zaug, et al., 1986, Nature, 324, 429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been and Cech, 1986, Cell, 47, 207-216).
  • Cech-type ribozymes such as the one that occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and that has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224,
  • the Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in the gene encoding the MSPI.
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells that express the MSPI in vivo.
  • a preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol JH or pol U promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous mRNA encoding the MSPI and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficacy.
  • Endogenous MSPI expression can also be reduced by inactivating or "knocking out" the gene encoding the MSPI, or the promoter of such a gene, using targeted homologous recombination (e.g., see Smithies, et al., 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51:503-512; Thompson et al., 1989, Cell 5:313-321; and Zijlstra et al., 1989, Nature 342:435-438, each of which is incorporated by reference herein in its entirety).
  • targeted homologous recombination e.g., see Smithies, et al., 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51:503-512; Thompson et al., 1989, Cell 5:313-321; and Zijlstra et al., 1989, Nature 342:435-438, each of which is incorporated by reference herein in its entirety).
  • a mutant gene encoding a non-functional MSPI (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous gene (either the coding regions or regulatory regions of the gene encoding the MSPI) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express the target gene in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the target gene.
  • Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive target gene (e.g., see Thomas and Capecchi, 1987 and Thompson, 1989, supra).
  • this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors.
  • the endogenous expression of a gene encoding an MSPI can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the gene (i.e., the gene promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene encoding the MSPI in target cells in the body.
  • deoxyribonucleotide sequences complementary to the regulatory region of the gene i.e., the gene promoter and/or enhancers
  • triple helical structures that prevent transcription of the gene encoding the MSPI in target cells in the body.
  • Nucleic acid molecules to be used in triplex helix formation for the inhibition of transcription should be single stranded and composed of deoxynucleotides.
  • the base composition of these oligonucleotides must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC+ triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules may be chosen that are purine-rich, for example, contain a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • the potential sequences that can be targeted for triple helix formation may be increased by creating a so called “switchback" nucleic acid molecule.
  • Switchback molecules are synthesized in an alternating 5 '-3', 3 '-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the technique may so efficiently reduce or inhibit the transcription (triple helix) or translation (antisense, ribozyme) of mRNA produced by normal gene alleles of an MSPI that the situation may arise wherein the concentration of MSPI present may be lower than is necessary for a normal phenotype.
  • gene therapy may be used to introduce into cells nucleic acid molecules that encode and express the MSPI that exhibitnormal gene activity and that do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments are being utilized.
  • Antisense RNA and DNA, ribozyme, and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules, as discussed above. These include techniques for chemically synthesizing oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule.
  • DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • the present invention also provides assays for use in drug discovery in order to identify or verify the efficacy of agents for treatment or prevention of MS.
  • Candidate agents can be assayed for their ability to restore MSF or MSPI levels in a subject having MS towards levels found in subjects free from MS or to produce similar changes in experimental animal models of MS.
  • Agents able to restore MSF or MSPI levels in a subject having MS towards levels found in subjects free from MS or to produce similar changes in experimental animal models of MS can be used as lead agents for further drug discovery, or used therapeutically.
  • MSF and MSPI expression can be assayed by the Preferred Technology, irnmunoassays, gel electrophoresis followed by visualization, detection of MSPI activity, or any other method taught herein or known to those skilled in the art.
  • Such assays can be used to screen candidate drugs, in clinical monitoring or in drug development, where abundance of an MSF or MSPI can serve as a surrogate marker for clinical disease.
  • in vitro assays can be carried out with cells representative of cell types involved in a subject's disorder, to determine if a agent has a desired effect upon such cell types.
  • Agents for use in therapy can be tested in suitable animal model systems prior to testing in humans, including but not limited to rats, mice, chickens, cows, monkeys, rabbits, etc.
  • suitable animal model systems prior to administration to humans, any animal model system known in the art may be used
  • animal models of MS include, but are not limited to, experimental autoimmune encephalomyelitis (EAE) (Steinman (1999) Neuron, 24:511-514).
  • EAE experimental autoimmune encephalomyelitis
  • transgenic animals can be produced with "knock-out" mutations of the gene or genes encoding one or more MSPIs.
  • a "knock-out" mutation of a gene is a mutation that causes the mutated gene to not be expressed, or expressed in an aberrant form or at a low level, such that the activity associated with the gene product is nearly or entirely absent.
  • the transgenic animal is a mammal, more preferably, the transgenic animal is a mouse.
  • candidate agents that modulate the expression of an MSPI are identified in non-human animals (e.g., mice, rats, monkeys, rabbits, and guinea pigs), preferably non-human animal models for MS, expressing the MSPI.
  • a candidate agent or a control agent is administered to the animals, and the effect of the candidate agent on expression of one or more MSPIs is determined.
  • a candidate agent that alters the expression of an MSPI can be identified by comparing the level of the selected MSPI or MSPIs (or mRNA(s) encoding the same)- in an animal or group of animals treated with a candidate agent with the level of the MSPIs(s) or mRNA(s) in an animal or group of animals treated with a control agent.
  • Techniques known to those of skill in the art can be used to determine the mRNA and protein levels, for example, in situ hybridization. The animals may or may not be sacrificed to assay the effects of a candidate agent.
  • candidate agents that modulate the activity of an MSPI or a biologically active portion thereof are identified in non-human animals (e.g., mice, rats, monkeys, rabbits, and guinea pigs), preferably non-human animal models for MS, expressing the MSPIs.
  • a candidate agent or a control agent is administered to the animals, and the effect of a candidate agent on the activity of an MSPI is determined.
  • a candidate agent that alters the activity of an MSPI can be identified by assaying animals treated with a control agent and animals treated with the candidate agent.
  • the activity of the MSPI can be assessed by detecting induction of a cellular second messenger of the MSPI (e.g., intracellular Ca2 + , diacylglycerol, IP3, etc.), detecting catalytic or enzymatic activity of the MSPI or binding partner thereof, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to an MSPI of the invention operably linked to a nucleic acid encoding a detectable marker, such as luciferase or green fluorescent protein), or detecting a cellular response (e.g., cellular differentiation or cell proliferation).
  • a reporter gene e.g., a regulatory element that is responsive to an MSPI of the invention operably linked to a nucleic acid encoding a detectable marker, such as luciferase or green fluorescent protein
  • a cellular response e.g., cellular differentiation or cell proliferation.
  • candidate agent that modulate the level or expression of an MSPI are identified in human subjects having MS, preferably those having severe MS.
  • a candidate agent or a control agent is administered to the human subject, and the effect of a candidate agent on MSPI expression is determined by analyzing the expression of the MSPI or the mRNA encoding the same in a biological sample (e.g., CSF, serum, plasma, or urine).
  • a candidate agent that alters the expression of an MSPI can be identified by comparing the level of the MSPI or mRNA encoding the same in a subject or group of subjects treated with a control agent to that in a subject or group of subjects treated with a candidate agent.
  • alterations in the expression of an MSPI can be identified by comparing the level of the MSPI or mRNA encoding the same in a subject or group of subjects before and after the administration of a candidate agent.
  • Techniques known to those of skill in the art can be used to obtain the biological sample and analyze the mRNA or protein expression.
  • the Preferred Technology described herein can be used to assess changes in the level of an MSPI.
  • candidate agents that modulate the activity of an MSPI are identified in human subjects having MS, preferably those having severe MS.
  • a candidate agent or a control agent is administered to the human subject, and the effect of a candidate agent on the activity of an MSPI is determined.
  • a test com candidate agent pound that alters the activity of an MSPI can be identified by comparing biological samples from subjects treated with a control agent to samples from subjects treated with the candidate agent.
  • alterations in the activity of an MSPI can be identified by comparing the activity of an MSPI in a subject or group of subjects before and after the administration of a candidate agent.
  • the activity of the MSPI can be assessed by detecting in a biological sample (e.g., CSF, serum, plasma, or urine) induction of a cellular signal transduction pathway of the MSPI (e.g., intracellular Ca2 + , diacylglycerol, JP3, etc.), catalytic or enzymatic activity of the MSPI or a binding partner thereof, or a cellular response, for example, cellular differentiation, or cell proliferation.
  • a biological sample e.g., CSF, serum, plasma, or urine
  • a cellular signal transduction pathway of the MSPI e.g., intracellular Ca2 + , diacylglycerol, JP3, etc.
  • catalytic or enzymatic activity of the MSPI or a binding partner thereof e.g., intracellular Ca2 + , diacylglycerol, JP3, etc.
  • a cellular response for example, cellular differentiation, or cell proliferation.
  • Techniques known to those of skill in the art can be used
  • a candidate agent that changes the level or expression of an MSPI towards levels detected in control subjects is selected for further testing or therapeutic use.
  • a candidate agent that changes the activity of an MSPI towards the activity found in control subjects is selected for further testing or therapeutic use.
  • candidate agent that reduce the severity of one or more symptoms associated with MS are identified in human subjects having MS, preferably subjects severe MS.
  • a candidate agent or a control agent is administered to the subjects, and the effect of a candidate agent on one or more symptoms of MS is dete ⁇ riined.
  • a candidate agent that reduces one or more symptoms can be identified by comparing the subjects treated with a control agent to the subjects treated with the candidate agent. Techniques known to physicians familiar with MS can be used to determine whether a candidate agent reduces one or more symptoms associated with MS.
  • a candidate agent that reduces clinical relapse rate, development of new MRI lesions, delays disability progression, enhances speed of clinical recovery, restores blood brain barrier status on MRI or delays increase of MRI lesion volume, protection of oligodendrocytes, axonal remyelination in a subject having MS will be beneficial for treating subjects having MS.
  • a candidate agent that reduces the severity of one or more symptoms associated with MS in a human having MS is selected for further testing or therapeutic use.
  • the invention provides methods of treatment (and prophylaxis) comprising administering to a subject an effective amount of an active agent.
  • An "active agent” as used herein comprises MSPIs, MSPI fragments, MSPI-related polypeptides, anti-MSPI antibodies, fragments of anti-MSPI antibodes and agents which modulate the expression of MSPIs e.g. agonists and antagonists of MSPIs.
  • the agent is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is preferably an animal and is preferably a mammal, and most preferably human.
  • compositions and methods of administration that can be employed when the agent comprises a nucleic acid are described above; additional appropriate formulations and routes of administration are described below.
  • a "pharmaceutical composition” as used herein comprises an active agent optionally with a pharmaceutically acceptable carrier.
  • a agent of the invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the agent, receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction can be enteral or parenteral and include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the pharmaceutical compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • compositions of the invention may be desirable to administer locally to the area in need of treatment; this may be achieved, for example, and not by way of limitation, by local infusion during surgery, topical application, e.g., by injection, by means of a catheter, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • administration can be by direct injection into CSF or at the site (or former site) of neurodegeneration or to CNS tissue.
  • the agent can be delivered in a vesicle, in particular a liposome (see Langer, 1990, Science 249:1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the agent can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, .1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drag Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., 1983, Macromol. S.ci. Rev. Macromol. Chem. 23:61 ; see also Levy et al., 1985, Science 228: 190; During et al., 1989,
  • a controlled release system can be placed in proximity of the therapeutic target, e.g. the CNS, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Other controlled release systems are discussed in the review by Langer (1990, Science
  • the agent of the invention is a nucleic acid encoding a protein
  • the nucleic acid can be administered in vivo as described supra.
  • compositions comprise a therapeutically effective amount of a agent, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences", Ed. E.W. Martin, ISBN: 0-912734-04-3, Mack Publishing Co.
  • Such compositions will contain a therapeutically effective amount of the agent, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be , mixed prior to administration.
  • the agents of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the agent of the invention which will be effective in the treatment of MS can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the active agent, the route of administration of the active agent, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active agent per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects (a) approval by the agency of manufacture, use or ⁇ sale for human administration, (b) directions for use, or both.
  • a protein assay (Pierce BCA Cat # 23225) was performed on each CSF sample as received. Prior to protein separation, each sample was processed for selective depletion of certain proteins, in order to enhance and simplify protein separation and facilitate analysis by removing proteins that may interfere with or limit analysis of proteins of interest. See WO 99/63351, which is inco ⁇ orated by reference in its entirety, with particular reference to pages 3 and 6.
  • CSF depletion Removal of albumin, haptoglobin, transferrin and immunoglobin G (IgG) from CSF ("CSF depletion") was achieved by an affinity chromatography purification step in which the sample was passed through a series of Hi-TrapTM columns containing immobilized antibodies for selective removal of albumin, haptoglobin and transferrin, and protein G for selective removal of immunoglobin G.
  • Hi-TrapTM columns Two affinity columns in a tandem assembly were prepared by coupling antibodies to protein G-sepharose contained in Hi-TrapTM columns (Protein G-Sepharose Hi-Trap columns (1 ml) Pharmacia Cat. No. 17-0404-01).
  • the chromatographic procedure was automated using an Akta Fast Protein Liquid Chromatography (FPLC) System such that a series of up to seven runs could be performed sequentially.
  • the samples were passed through the series of 3 Hi-Trap columns in which the affinity chromatography media selectively bind the above proteins thereby removing them from the sample.
  • Fractions typically 3 ml per tube
  • Flowthrough fractions unbound material
  • Bound/Eluted fractions bound proteins
  • the eluate containing unbound material was collected in fractions which were pooled, desalted/concentrated by centrifugal ultrafiltration and stored to await further analysis by 2D PAGE.
  • a volume of depleted CSF containing approximately 100-150 ⁇ g of total protein was aliquoted and an equal volume of 10% (w/v) SDS (Fluka 71729), 2.3% (w/v) dithiothreitol (BDH 443852A) was added.
  • the sample was heated at 95°C for 5 rnins, and then allowed to cool to 20°C. 125 ⁇ l of the following buffer was then added to the sample:
  • JEF Isoelectric focusing
  • the current limit was set to 10mA for 12 gels, and the wattage limit to 5W.
  • the temperature was held at 20°C throughout the run.
  • the strips were immediately removed and immersed for 10 rnins at 20°C in a first solution of the following composition: 6M urea; 2% (w/v) DTT; 2% (w/v) SDS; 30% (v/v) glycerol (Fluka 49767); 0.05M Tris HCl, pH 6.8 (Sigma Cat T-1503).
  • the strips were removed from the first solution and immersed for 10 mins at 20°C in a second solution of the following composition: 6M urea; 2% (w/v) iodoacetamide (Sigma 1-6125); 2% (w/v) SDS; 30% (v/v) glycerol; 0.05M Tris/HCl, pH 6.8.
  • the strips were loaded onto supported gels for SDS-PAGE according to Hochstrasser et al., 1988, Analytical Biochemistry 173: 412-423 (inco ⁇ orated herein by reference in its ' entirety), with modifications as specified below.
  • the gels were cast between two glass plates of the following dimensions: 23cm wide x 24cm long (back plate); 23cm wide x 24cm long with a 2cm deep notch in the central 19cm (front plate).
  • the back plate was treated with a 0.4% solution of g-methacryl-oxypropyltrimethoxysilane in ethanol (BindSilanej; Pharmacia Cat. # 17-1330-01).
  • the front plate was treated with a 2% solution of dimethyldichlorosilane dissolved in octamethyl cyclo-octasilane (RepelSilanej Pharmacia Cat. # 17-1332-01) to reduce adhesion of the gel.
  • a 9-16% linear polyacrylamide gradient was cast, extending up to a point 2cm below the level of the notch in the front plate, using the Angelique gradient casting system CLarge Scale Biology).
  • Stock solutions were as follows. Acrylamide (40% in water) was from Serva (Cat. # 10677). The cross-linking agent was PDA (BioRad 161-0202), at a concentration of 2.6% (w/w) of the total starting monomer content.
  • the gel buffer was 0.375M Tris HCl, pH 8.8.
  • the polymerization catalyst was 0.05% (v/v) TEMED (BioRad 161-0801), and the initiator was 0.1% (w/v) APS (BioRad 161-0700). No SDS was included in the gel and no stacking gel was used. The cast gels were allowed to polymerize at 20°C overnight, and then stored at 4°C in sealed polyethylene bags with 6ml of gel buffer, and were used within 4 weeks.
  • a solution of 0.5% (w/v) agarose (Fluka Cat 05075) was prepared in running buffer (0.025M Tris, 0.198M glycine (Fluka 50050), 1% (w/v) SDS, supplemented by a trace of bromophenol blue).
  • the agarose suspension was heated to 70°C with stirring, until the agarose had dissolved.
  • the top of the supported 2nd D gel was filled with the agarose solution, and the equilibrated strip was placed into the agarose, and tapped gently with a palette knife until the gel was intimately in contact with the 2nd D gel.
  • the gels were placed in the 2nd D running tank, as described by Amess et al., 1995, Electrophoresis 16: 1255-1267 (inco ⁇ orated herein by reference in its entirety).
  • the tank was filled with running buffer (as above) until the level of the buffer was just higher than the top of the region of the 2nd D gels which contained polyacrylamide, so as to achieve efficient cooling of the active gel area.
  • Running buffer was added to the top buffer compartments formed by the gels, and then voltage was applied immediately to the gels using a Consort E-833 power supply. For 1 hour, the gels were ran at 20mA/gel.
  • the wattage limit was set to 150W for a tank containing 6 gels, and the voltage limit was set to 600V.
  • the gels were immediately removed from the tank for fixation.
  • the top plate of the gel cassette was carefully removed, leaving the gel bonded to the bottom plate.
  • the bottom plate with its attached gel was then placed into a staining apparatus, which can accommodate 12 gels.
  • the gels were completely immersed in fixative solution of 40% (v/v) ethanol (BDH 28719), 10% (v/v) acetic acid (BDH 100016X), 50% (v/v) water (MilliQ-Millipore), which was continuously circulated over the gels.
  • the fixative was drained from the tank, and the gels were primed by immersion in 7.5% (v/v) acetic acid, 0.05% (w/v) SDS, 92.5% (v/v) water for 30 rnins.
  • the priming solution was then drained, and the gels were stained by complete immersion for 4 hours in a staining solution of Pyridinium, 4-[2-[4-(dipentylamino)-2-trifluoromethylphenyl] ethenyl]-l-(sulfobutyl)-, inner salt, prepared by diluting a stock solution of this dye (2mg/ml in DMSO) in 7.5% (v/v) aqueous acetic acid to give a final concentration of 1.2 mg/1; the staining solution was vacuum filtered through a 0.4 ⁇ m filter (Duropore) before use.
  • a staining solution of Pyridinium, 4-[2-[4-(dipentylamino)-2-trifluoromethylphenyl] ethenyl]-l-(sulfobutyl)-, inner salt prepared by diluting a stock solution of this dye (2mg/ml in DMSO) in 7.5% (v/v)
  • a computer-readable output was produced by imaging the fluorescently stained gels with the preferred scanner (Oxford Glycosciences, Oxford, UK described in section 5.2, supra.
  • This scanner has a gel carrier with four integral fluorescent markers (Designated Ml, M2, M3, M4) that are used to correct the image geometry and are a quality control feature to confirm that the scanning has been performed correctly.
  • the gels were removed from the stain, rinsed with water and allowed to air dry briefly, and imaged on the preferred scanner. After imaging, the gels were sealed in polyethylene bags containing a small volume of staining solution, and then stored at 4°C.
  • the output from the scanner was first processed using the MELANIE7 U 2D PAGE analysis program (Release 2.2, 1997, BioRad Laboratories, Hercules, California, Cat. #
  • Landmark identification was used to determine the pi and MW of features detected in the images. Twelve landmark features, designated CSFl to CSF12, were identified in a standard CSF image obtained from a pooled sample. These landmark features are identified in Figure 1 and were assigned the pi and/or MW values identified in Table X.
  • each feature in the study gels was then assigned a pi value by linear inte ⁇ olation or extrapolation (using the MELANIE7- ⁇ software) to the two nearest landmarks, and was assigned a MW value by linear inte ⁇ olation or extrapolation (using the MELANJE7- ⁇ software) to the two nearest landmarks.
  • Images were edited to remove gross artifacts such as dust, to reject images which had gross abnormalities such as smearing of protein features, or were of too low a loading or overall image intensity to allow identification of more than the most intense features, or were of too poor a resolution to allow accurate detection of features. Images were then compared by pairing with one common image from the whole sample set. This common image, the "primary master image", was selected on the basis of protein load (maximum load consistent with maximum feature detection), a well resolved myoglobin region, (myoglobin was used as an internal standard), and general image quality. Additionally, the primary master image was chosen to be an image which appeared to be generally representative of all those to be included in the analysis.
  • Each of the remaining study gel images was individually matched to the primary master image such that common protein features were paired between the primary master image and each individual study gel image as described below.
  • each study gel was adjusted for maximum alignment between its pattern of protein features, and that of the primary master, as follows.
  • Each of the study gel images was individually transformed into the geometry of the primary master image using a multi-resolution wa ⁇ ing procedure. This procedure corrects the image geometry for the distortions brought about by small changes in the physical parameters of the electrophoresis separation process from one sample to another. The observed changes are such that the distortions found are not simple geometric distortions, but rather a smooth flow, with variations at both local and global scale.
  • the fundamental principle in multi-resolution modeling is that smooth signals may be modeled as an evolution through 'scale space', in which details at successively finer scales are added to a low resolution approximation to obtain the high resolution signal.
  • This type of model is applied to the flow field of vectors (defined at each pixel position on the reference image) and allows flows of arbitrary smoothness to be modeled with relatively few degrees of freedom.
  • Each image is first reduced to a stack, or pyramid, of images derived from the initial image, but- smoothed and reduced in resolution by a factor of 2 in each direction at every level (Gaussian pyramid) and a corresponding difference image is also computed at each level, representing the difference between the smoothed image and its progenitor (Laplacian pyramid).
  • the Laplacian images represent the details in the image at different scales.
  • the wa ⁇ ing process brought about good alignment between the common features in the primary master image, and the images for the other samples.
  • the MELANEE7 U 2D PAGE analysis program was used to calculate and record approximately 500-700 matched feature pairs between the primary master and each of the other images.
  • the accuracy of this program was significantly enhanced by the alignment of the images in the manner described above.
  • all pairings were finally exarnined by eye in the MelView interactive editing program and residual recognizably incorrect pairings were removed.
  • the gel selected to be the primary master gel was judged to be insufficiently representative of the study gels to serve as a primary master gel. In that case, the gel chosen as the primary master gel was rejected, and different gel was selected as the primary master gel, and the process was repeated.
  • a composite master image was thus generated by initialising the primary master image with its feature descriptors. As each image was transformed into the primary master geometry, it was digitally summed pixel by pixel into the composite master image, and the features that had not been paired by the procedure outlined above were likewise added to the composite master image description, with their centroids adjusted to the master geometry using the flow field correction.
  • MCI molecular cluster index
  • An MCI identifies a set of matched features on different images.
  • an MCI represents a protein or proteins eluting at equivalent positions in the 2D separation in different samples.
  • LEVIS Information Management System
  • a second non-overlapping selection strategy is based on qualitative presence or absence alone. Using this procedure, a percentage feature presence was calculated across the control samples and MS samples for each MCI which was a potential MSF based on such qualitative criteria alone, i.e. presence or absence. The MCIs which recorded a percentage feature presence of 95% or more on MS samples and a percentage feature presence of 15% or less on control samples, were selected as the qualitative differential MSFs with 85% selectivity. A second group of qualitative differential MSFs with 85% selectivity were formed by those MCIs which recorded a percentage feature presence of 95% or more on control samples and a percentage feature presence of 15% or less on MS samples.
  • a third non-overlapping selection strategy is based on the fold change.
  • a fold change representing the ratio of the average normalized protein abundances of the MSFs within an MCI, was calculated for each MCI between each set of controls and MS samples.
  • a 95% confidence limit for the mean of the fold changes was calculated.
  • the MCIs with fold changes which fall outside the confidence limit were selected as MSFs which met the criteria of the significant fold change threshold with 95% selectivity. Because the MCI fold changes are based on a 95% confidence limit, it follows that the significant fold change threshold is itself 95%.
  • MSFs to be selected on the basis of: (a) statistical significance as measured by the Wilcoxon Rank-Sum test, (b) qualitative differences with a chosen selectivity, or (c) a significant fold change threshold with a chosen selectivity,
  • Tryptic peptides were analyzed by mass spectrometry using a PerSeptive Biosystems Voyager- DETM STR Matrix-Assisted Laser Deso ⁇ tion Ionization Time-of-Flight (MALDI-TOF) mass spectrometer, and selected tryptic peptides were analyzed by tandem mass spectrometry (MS/MS) using a Micromass Quadrupole Time-of-Flight (Q-TOF) mass spectrometer (Micromass, Altrincham, U.K.) equipped with a nanoflowTM electrospray Z-spray source.
  • MALDI-TOF PerSeptive Biosystems Voyager- DETM STR Matrix-Assisted Laser Deso ⁇ tion Ionization Time-of-Flight
  • MS/MS tandem mass spectrometry
  • Q-TOF Micromass Quadrupole Time-of-Flight
  • the following example illustrates the use of an MSPI of the invention for screening or diagnosis of MS, determining the prognosis of a MS patient, or monitoring the effectiveness of MS therapy.
  • the following example also illustrates the use of modulators (e.g., agonists or antagonists) of an MSPI of the invention to treat or prevent MS.
  • Cathepsins are part of the papain superfamily of lysosomal cysteine proteinases that play a major role in intracellular protein degradation (Kirschke, H. and Barrett, A.J. (1987) in Lysosomes: Their role in protein breakdown (Glaumann, H., and Ballard, F.J., eds) 193-238, Academic Press, London).
  • Cathepsin S appears to be essential for antigen presentation by major histocompatibility complex (MHC) class TL molecules.
  • Cathepsin may have putative roles in several pathologies including cancer (Hughes et al. (1998) Proc. Nat. Acad. Sci. 95: 12410-12415) and neuronal cell death (Isahara et. Al (1999) Neuroscience 91:233-249), such as Cathepsin B in post ischaemic hippocampal neuronal death (Yamashima et al. (1998) Eur J Neurosci 5:1723-33).
  • Cathepsin L may participate in various pathological processes including arthritis (Esser et al.
  • Cathepsin L is synthesized as an inactive proenzyme, which is subsequently processed to the mature form through cleavage of an amino-terminal proregion, located between the signal sequence and the N terminus of the mature enzyme (Menar et al. (1998) J Biol Chem 273:4478-4484), resulting in isoforms.
  • the proregion may regulate catalytic activity, and has been shown to be a potent inhibitor of the mature enzyme (Carmona et al. (1996) Biochemistry 35:8149-8157).
  • Cathepsin L with a molecular weight of 40,820 Da and pi of 4.37 has been shown herein to be significantly increased in the cerebrospinal fluid (CSF) of subjects having MS as compared with the CSF of subjects free from MS (see Table ⁇ - MSPI-300).
  • CSF cerebrospinal fluid
  • quantitative detection of Cathepsin L in CSF can be used to diagnose MS, determine the progression of MS or monitor the effectiveness of a therapy for MS.
  • agents that modulate e.g., downregulate
  • the expression, activity or both the expression and activity of Cathepsin L are administered to a subject in need of treatment or for prophylaxis of MS.
  • Antibodies that modulate the expression, activity or both the expression and activity of Cathepsin L are suitable for this pu ⁇ ose.
  • nucleic acids coding for all or a portion of Cathepsin L, or nucleic acids complementary to all or a portion of Cathepsin L may be administered.
  • Cathepsin L, or fragments of the Cathepsin L polypeptide may also be administered.
  • the invention also provides screening assays to identify additional agents that modulate the expression of
  • Cathepsin L or activity of Cathepsin L Agents that modulate the expression of Cathepsin L in vitro can be identified by comparing the expression of Cathepsin L in cells treated with a candidate agent to the expression of Cathepsin L in cells treated with a control agent (e.g., saline).
  • Methods for detecting expression of Cathepsin L are known in the art and include measuring the level of Cathepsin L RNA (e.g., by northern blot analysis or RT-PCR) and measuring Cathepsin L protein (e.g., by immunoassay or western blot analysis).
  • Agents that modulate the activity of Cathepsin L can be identified by comparing the ability of a candidate agent to agonize or antagonize a function of Cathepsin L, such as its neurotrophic or neuroprotective activity, to the ability of a control agent (e.g., saline) to inhibit the same function of Cathepsin L.
  • Agents capable of modulating Cathepsin L activity are identified as agents suitable for further development as a agent useful for the treatment of MS.
  • candidate agents may be tested for their ability to antagonize the activity of Cathepsin L.
  • Cathepsin L Agents identified in vitro that affect the expression or activity of Cathepsin L can be tested in vivo in animal models of MS, or in subjects having MS, to determine their therapeutic efficacy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Microbiology (AREA)
  • Neurosurgery (AREA)
  • Public Health (AREA)
  • Toxicology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • Rehabilitation Therapy (AREA)
  • Rheumatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)

Abstract

La présente invention concerne des procédés et des compositions permettant le criblage, le diagnostic et la prévision de la sclérose en plaques, le suivi de l'efficacité du traitement de la sclérose en plaques, l'identifications des patients les plus susceptibles de répondre à un traitement thérapeutique particulier, et la mise au point de médicaments. Cette invention concerne également les caractéristiques associées à la sclérose en plaques (MSFs) pouvant être détectées par une électrophorèse tridimensionnelle de fluides corporels tels que le liquide céphalo-rachidien. L'invention a également pour objet des isoformes de la protéine associée à la sclérose en plaques (MSPIs) pouvant être détectées dans des fluides corporels tels que le liquide céphalo-rachidien, des préparations combinant des MSPIs isolées, des anticorps immunospécifiques de MSPIs et des kits comprenant les éléments mentionnés ci-dessus.
EP02715572A 2001-01-26 2002-01-25 Diagnostic et traitement de la sclerose en plaques Withdrawn EP1354199A2 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US26440401P 2001-01-26 2001-01-26
US264404P 2001-01-26
US33164701P 2001-11-20 2001-11-20
US331647P 2001-11-20
PCT/GB2002/000330 WO2002059604A2 (fr) 2001-01-26 2002-01-25 Proteines, genes et leur utilisation pour le diagnostic et le traitement de la sclerose en plaques

Publications (1)

Publication Number Publication Date
EP1354199A2 true EP1354199A2 (fr) 2003-10-22

Family

ID=26950517

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02715572A Withdrawn EP1354199A2 (fr) 2001-01-26 2002-01-25 Diagnostic et traitement de la sclerose en plaques

Country Status (4)

Country Link
EP (1) EP1354199A2 (fr)
JP (1) JP2004532386A (fr)
AU (1) AU2002225219A1 (fr)
WO (1) WO2002059604A2 (fr)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5048201B2 (ja) 2000-08-04 2012-10-17 ディーエムアイ バイオサイエンシズ インコーポレイテッド ジケトピペラジン類の使用方法とジケトピペラジン類を含む組成物
US7015004B2 (en) * 2001-11-23 2006-03-21 Syn X Pharma, Inc. Inter-alpha trypsin inhibitor biopolymer marker indicative of insulin resistance
US20030113808A1 (en) * 2001-12-13 2003-06-19 George Jackowski Apolipoprotein biopolymer markers predictive of alzheimers disease
CA2527916A1 (fr) * 2003-03-14 2004-09-30 Aaron B. Kantor Marqueurs biologiques pour le diagnostic de l'arthrite rhumatoide
EP2517721A3 (fr) 2003-05-15 2013-04-10 DMI Biosciences, Inc. Traitement de maladies à mediation des lymphocytes T
US9408891B2 (en) 2003-11-12 2016-08-09 The Trustees Of The University Of Pennsylvania Methods of using gelsolin to treat or prevent bacterial sepsis
CA2860173C (fr) 2004-05-12 2021-12-14 The Brigham And Women's Hospital, Inc. Determination du niveau de gelsoline dans le plasma en vue de predire le risque de developpement d'une infection
US20050266467A1 (en) * 2004-05-19 2005-12-01 Ppd Biomarker Discovery Sciences, Llc Biomarkers for multiple sclerosis and methods of use thereof
WO2006005583A2 (fr) * 2004-07-12 2006-01-19 Geneprot Inc. Especes de polypeptides secretes impliques dans la sclerose en plaques
WO2006074787A2 (fr) * 2004-12-27 2006-07-20 Geneprot Inc. Espece de polypeptides utiles pour le traitement de troubles neurologiques
WO2006069739A2 (fr) * 2004-12-27 2006-07-06 Geneprot Inc. Polypeptide utile pour traiter des troubles neurologiques
US20070017809A1 (en) * 2005-07-21 2007-01-25 Power3 Medical Products, Inc. Assay for ALS and ALS-like disorders
PT2002258T (pt) * 2006-03-15 2017-12-18 The Brigham And Women`S Hospital Inc Utilização de gelsolina para o tratamento da esclerose múltipla e para o diagnóstico de doenças neurológicas
JP5322917B2 (ja) * 2006-03-15 2013-10-23 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド 炎症性疾患の診断および処置のためのゲルゾリンの使用
JPWO2009020058A1 (ja) * 2007-08-03 2010-11-04 学校法人慶應義塾 脱髄病変へのドラッグデリバリーシステム及び脱髄病変の生化学的マーカー
AU2013276991C1 (en) * 2007-08-13 2016-08-11 Takeda Pharmaceutical Company Limited IVIG Modulations of Chemokines for Treatment of Multiple Sclerosis, Alzheimer's Disease, and Parkinson's Disease
ES2477271T3 (es) * 2007-08-13 2014-07-16 Baxter International Inc. Modulación por IVIG de quimioquinas para el tratamiento de la esclerosis múltiple, la enfermedad de Alzheimer y la enfermedad de Parkinson
ES2531827T3 (es) 2008-01-25 2015-03-20 The General Hospital Corporation Usos terapéuticos de gelsolina en insuficiencia renal
US8217047B2 (en) 2008-05-27 2012-07-10 Dmi Acquisition Corp. Therapeutic methods and compounds
WO2010005387A1 (fr) * 2008-07-10 2010-01-14 Astrazeneca Ab Procédé et marqueurs inédits pour le diagnostic de la sclérose en plaques
EP2613786A4 (fr) 2010-09-07 2013-10-23 Dmi Acquisition Corp Traitement de maladies
EP3101426B1 (fr) * 2010-12-16 2019-05-15 National Institute of Advanced Industrial Science And Technology Procédé d'enrichissement et de séparation de fluide spinal glycoprotéine, procédé de recherche de marqueur pour les maladies du système nerveux central qui utilise le procédé susmentionné et marqueur pour les maladies du système nerveux central
EP3721884A1 (fr) 2011-10-10 2020-10-14 Ampio Pharmaceuticals, Inc. Traitement de maladies articulaires dégénératives avec da-dkp (= diketopiperazine aspartyl-alanyl)
MX2014003856A (es) 2011-10-10 2015-01-16 Ampio Pharmaceuticals Inc Dispositivos medicos implantables con tolerancia inmune incrementada y metodos para hacerlos e implantarlos.
EP2771007B1 (fr) 2011-10-28 2018-04-04 Ampio Pharmaceuticals, Inc. Traitement de la rhinite
ITMI20120865A1 (it) * 2012-05-18 2013-11-19 Antonio Bertolotto Biomarcatori per patologie del sistema nervoso centrale
US9808454B2 (en) 2013-03-15 2017-11-07 Ampio Pharmaceuticals, Inc. Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
KR20170045274A (ko) 2014-08-18 2017-04-26 앰피오 파마슈티컬스 인코퍼레이티드 관절 징후의 치료
WO2016209969A1 (fr) 2015-06-22 2016-12-29 Ampio Pharmaceuticals, Inc. Utilisation de fractions d'albumine de sérum humain de bas poids moléculaire pour traiter les maladies
RU2694614C1 (ru) * 2019-01-22 2019-07-17 Общество с ограниченной ответственностью "ВЕСТТРЭЙД ЛТД" (ООО "ВЕСТТРЭЙД ЛТД") Способ определения активности патологического процесса у больных рассеянным склерозом
WO2020172262A1 (fr) * 2019-02-19 2020-08-27 James Janine Composition à base de chrome et procédés associés

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL137836A0 (en) * 1998-02-13 2001-10-31 Oxford Glycosciences Uk Ltd Methods and compositions for diagnosis of hepatoma

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO02059604A2 *

Also Published As

Publication number Publication date
WO2002059604A3 (fr) 2003-07-03
WO2002059604A2 (fr) 2002-08-01
WO2002059604A8 (fr) 2002-11-21
AU2002225219A1 (en) 2002-08-06
JP2004532386A (ja) 2004-10-21

Similar Documents

Publication Publication Date Title
WO2002059604A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la sclerose en plaques
US20040053309A1 (en) Proteins, genes and their use for diagnosis and treatment of kidney response
US20070015217A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20040110938A1 (en) Proteins, genes and their use for diagnosis and treatment of schizophrenia
EP1408333A2 (fr) Diagnostic et traitement de la maladie d'Alzheimer
EP1384079A2 (fr) Proteines, genes et leur utilisation dans le cadre du diagnostic et du traitement du cancer du sein
US20030064411A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20030152935A1 (en) Proteins, genes and their use for diagnosis and treatment of breast cancer
EP1379879A2 (fr) Diagnostic et traitement de la maladie d'alzheimer
US20020142303A1 (en) Proteins, genes and their use for diagnosis and treatment of Schizophrenia
EP1159618A1 (fr) Proteines permettant le diagnostic et le traitement du cancer du sein
EP1259819A2 (fr) Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires
US20030032200A1 (en) Proteins, genes and their use for diagnosis and treatment of bipolar affective disorder (BAD) and unipolar depression
EP1259818A2 (fr) Diagnostic et traitement de la schizophrenie
US20040203022A1 (en) Proteins and genes for diagnosis and treatment of ErbB2-related cancer
WO2001069261A2 (fr) Proteines, genes et leur utilisation dans le diagnostic et le traitement de la demence vasculaire
US20040022794A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20040043425A1 (en) Proteins, genes and their use for diagnosis and treatment of chronic asthma
US20030077603A1 (en) Methods and compositions for diagnosis of hepatoma
US20030092197A1 (en) Proteins, genes and their use for diagnosis and treatment of cardiac response
WO2002054080A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse vasculaire
WO2002054079A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse cardiaque

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030722

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050802