WO2010015834A1 - Antibodies specific for misfolded proteins and methods for their production - Google Patents

Antibodies specific for misfolded proteins and methods for their production Download PDF

Info

Publication number
WO2010015834A1
WO2010015834A1 PCT/GB2009/001944 GB2009001944W WO2010015834A1 WO 2010015834 A1 WO2010015834 A1 WO 2010015834A1 GB 2009001944 W GB2009001944 W GB 2009001944W WO 2010015834 A1 WO2010015834 A1 WO 2010015834A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
disease
prp
target antigen
antigen
Prior art date
Application number
PCT/GB2009/001944
Other languages
French (fr)
Inventor
Mourad Tayebi
Original Assignee
The Royal Veterinary College
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 The Royal Veterinary College filed Critical The Royal Veterinary College
Publication of WO2010015834A1 publication Critical patent/WO2010015834A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2872Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against prion molecules, e.g. CD230
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/32Immunoglobulins specific features characterized by aspects of specificity or valency specific for a neo-epitope on a complex, e.g. antibody-antigen or ligand-receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues

Definitions

  • the present application relates to novel antibodies and to methods for producing antibodies, in particular antibodies capable of binding to prion proteins.
  • Transmissible spongiform encephalopathies or prion diseases comprise a group of invariably fatal neurodegenerative disorders affecting humans and animals, characterised clinically in humans by a rapidly progressive dementia i.e. memory loss and cognitive deficit and neuropathologically, by spongiform degeneration.
  • Prion diseases such as CJD, BSE, together with Alzheimer's disease and certain forms of Parkinsonism are examples of disorders caused by protein misfolding and in prion disease, an abnormally folded protein (called prions or PrP ), derived from a normal cell surface protein (called PrP ), accumulates in the brain and other organs of affected animals. PrP and PrP are identical when denatured, meaning that the toxicity and infectivity of PrP Sc is wholly dependent on its conformation, i.e. how it is folded.
  • the human prion diseases include kuru, Creutzfeldt-Jacob disease (CJD), Gerstmann- Straussler-Scheinker syndrome (GSS), sporadic and familial fatal insomnia (FFI).
  • GSS, FFI and 10-15% of CJD are dominantly inherited disorders associated with specific mutations in the PRNP gene l , whereas 85-90% of CJD occur sporadically, and are not associated with mutations in the human prion protein gene .
  • Iatrogenic CJD has resulted from the use of contaminated neurosurgical instruments, corneal grafts, dura matter grafts, and pituitary derived hormone treatments.
  • scrapie In animals, the prototypic naturally occurring prion disease is scrapie, which affects sheep and goats. Scrapie has been recognised for over 200 years 3 and has a worldwide distribution. Other and more recently recognised animal diseases include transmissible mink encephalopathy, chronic wasting disease of mule, deer, and elk 4 , and bovine spongiform encephalopathy (BSE) 5 . Prion diseases are characterised by their transmissibility to a wide range of experimental animal species, including mice, hamsters, rats, and demonstrated by the transmission of kuru to chimpanzees 6 , and CJD and GSS to primates 7 8 .
  • Prion diseases can be transmitted by intracranial inoculation for experimental purposes but the appearance in UK cattle in 1986 of BSE, which rapidly evolved into a major epidemic 9;i0 , was widely attributed to transmission of sheep scrapie to cattle via contaminated feed prepared from rendered carcasses 9 .
  • the more recently described feline spongiform encephalopathy of domestic cats u and spongiform encephalopathies of a number of zoo animals, the so-called exotic ungulate encephalopathies ' are also recognised as animal prion diseases, and are thought to have resulted from the same contaminated food given to cattle.
  • a need for testing slaughtered cattle over 30 months of age for bovine spongiform encephalopathy has increased the momentum for developing rapid diagnostic tests. However, perhaps the greatest need is for tests with applicability in the sub-clinical phase of prion disease, requiring a specificity that is currently beyond available immunodiagnostic methods.
  • spongiform vacuolation astrocytosis
  • neuronal loss a progressive neuronal loss.
  • the spongiform change is not seen by the naked eye and vacuoles measure 1 to 50 ⁇ m in diameter.
  • Proliferation of astrocytic glial cells is another classical marker of prion diseases. This usually accompanies a loss of nerve cells in the surrounding areas of the brain.
  • Amyloid plaques are deposited in the brains of about 15% of prion disease cases and demonstrate the characteristic red/green birefringence when stained with congo red and exposed to polarised light.
  • prion rods were also purified then used to produce anti-PrP antibodies in both rabbits 22 and mice 23 . Similar to the SAF preparations, prion rods also undergo substantial and stringent purification steps 22 which could lead to their denaturation and increased immunogenecity while leading to a remarkable reduction in infectivity.
  • the peptide vaccination approach has proven to be a useful strategy for generating PrP Sc - selective antibodies ' °, although it is proving crucial to further investigate the structure and mechanisms of formation of PrP Sc and be able to demonstrate clearly whether these immunogenic motifs are prion disease-associated and not a shared generic epitope with other amyloid-forming diseases .
  • mice were immunised with DNA 3 or RNA 43 coding for different human prion proteins, following the synthesis of vectors expressing individual genotypes of either the cellular prion gene (PRNP) or mutant forms of the gene.
  • PRNP cellular prion gene
  • Full-length recombinant bovine PrP 38 (rbPrP 23"231 ), recombinant murine 4 ⁇ 6 (rmPrP 23'231 ), and recombinant ovine PrP 47 (roPrP 23 231 ) as well as truncated recombinant human PrP 48 (rhPrP 91"231 ) can be expressed in Escherichia coli as an ⁇ isoform which can then be folded into ⁇ isoforms after reduction of the disulfide bond and lowering the pH to 4.0.
  • the 15B3 49 antibody raised against rbPrP 23"231 appears to be selective for the disease-isoform of PrP, although no further application of this antibody has been reported.
  • Monoclonal antibodies to PrP have been successfully raised by various methods in Prn-p 0/0 mice 50 that do not express prion protein 23>38 - 51 53 .
  • generation of monoclonal antibodies that bind specifically the native conformation PrP Sc has been difficult to produce.
  • an IgG2a kappa chain monoclonal antibody 21 was produced by immunising C57BL/6J mice with SAF derived from the hamster 263K prion strain and reacted with hamster but not mouse SAF-derived 263K PrP, indicating strong influence of the host species in determining the antigenic response by the existence of prion epitopes that are species specific.
  • 3F4 was tested with homogenates from normal brain tissues of different species and reacted with both hamster and human PrP in brain preparations but failed to detect PrP in brain tissues from mouse, rat, rabbit, bovine, and sheep. Furthermore, pep-scan analysis of 3F4 has shown that it recognises an epitope present on hamster and human PrP between amino acids 109-113 which is dependent upon the presence of methionines at codon 109 and 112, lacking in other species tested. 3F4 has little or no affinity for native PrP Sc .
  • a method for producing an antibody specific for a target antigen comprising:-
  • the target antigen is a misfolded protein, more preferably a misfolded prion protein, further preferably PrP Sc or PrP c .
  • the target antigen is misfolded native PrP So .
  • the stabilising agent is selected from Congo red and Thiofiavin T.
  • step (ii) comprises incubating the stabilising agent with the target antigen for up to about 24 hours, preferably up to about 12 hours, further preferably between about 30 minutes and about 5 hours, more preferably about 1 hour and about 3 hours, most preferably about 1 hour or about 2 hours.
  • step (ii) comprises incubating the stabilising agent with the target antigen with continuous rotation.
  • the stabilising agent is incubated with the target antigen at about room temperature.
  • the stablising agent is present at a concentration of between about 50 ⁇ g/ml and about 1000 ⁇ g/ml, more preferably between about 100 ⁇ g/ml and about 500 ⁇ g/ml.
  • the concentration of stabilising agent is increased over time during step (ii), preferably from about 50 ⁇ g/ml to about 1000 ⁇ g/ml, most preferably from about 100 ⁇ g/ml to about 500 ⁇ g/ml.
  • the concentration of stabilising agent is increased continually throughout the incubation time of step (ii). In other embodiments, the concentration of stabilising agent is increased in stepped increments over the incubation time.
  • the sample containing the target antigen is a sample of animal tissue, further preferably spleen or a peripheral blood mononuclear cell (PBMC) or part of the CNS, eg. brain.
  • PBMC peripheral blood mononuclear cell
  • the animal tissue is homogenised prior to addition of the stabilising agent.
  • the target antigen is treated with a proteinase specific for non-target antigen proteins, prior to step (i), (ii) or (iii).
  • the proteinase is proteinase K.
  • the proteinase is present at a concentration of between about 10 ⁇ g/ml and about 100 ⁇ g/ml, more preferably about 50 ⁇ g/ml.
  • the target antigen is treated with the proteinase for up to about 2 hours, more preferably for up to about 1 hour, most preferably for about 30 minutes.
  • the target antigen is treated with the proteinase at a temperature of about 37°C.
  • the animal tissue is from an animal infected with a disease and wherein the target antigen is an indicator or a causative agent of that disease.
  • the disease is selected from a disease characterised by protein misfolding, preferably selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis.
  • prion diseases are selected from CJD, vCJD, BSE, GSS, FFI and Scrapie.
  • the conventional antibody is immobilised on a solid support by saturating a solid support with the conventional antibody.
  • the conventional antibody is immobilised on the solid support via a second antibody which is immobilised on the solid support and which binds the conventional antibody.
  • the host is selected from a mouse, camelid, rabbit, rat, sheep and cow.
  • the camelid is selected from an alpaca, camel and llama.
  • the complex is isolated from the sample.
  • the solid support is a bead, further preferably an immunomagnetic bead, more preferably selected from an IgG immunomagnetic bead, IgA immunomagnetic bead, protein G immunomagnetic bead, protein A immunomagnetic bead and streptavidin immunomagnetic bead.
  • the target antigen is immobilised on the solid support by incubating the target antigen with the solid support for up to about 24 hours, preferably for up to about 12 hours, further preferably between about 30 minutes and about 5 hours, more preferably about 1 hour and about 3 hours, most preferably about 1 hour or about 2 hours.
  • the target antigen is incubated with between about 10 5 and about 10 7 immunomagnetic particles, more preferably about 10 6 (25 ⁇ l) immunomagnetic particles.
  • the target antigen is incubated with the solid support at between about 0 0 C and about 25°C, preferably between about 2 0 C and about 8°C, more preferably at about 4°C or about room temperature.
  • step (ii) immobilising Congo Red bound PrP on an anti-PrP immunomagnetic bead to form a PrP/Congo Red immunomagnetic bead complex
  • step (ii) immunising a host with the PrP/Congo Red immunomagnetic bead complex
  • step (i) isolating antibodies specific for PrP Sc from the host.
  • the scrapie infected brain homogenate is incubated with proteinase K.
  • the scrapie infected brain homogenate is incubated with about 50 ⁇ g/ml proteinase K at about 37°C for about 30 minutes.
  • an antibody produced by a method of the invention.
  • the antibody is a monoclonal antibody.
  • the antibody is an antibody fragment.
  • the antibody is conjugated to a growth inhibitory agent.
  • the antibody is conjugated to a cytotoxic agent, for example a toxin, antibiotic, lytic enzyme or radioactive isotope.
  • the antibody is conjugated to a detectable marker, for example a fluorescent marker.
  • Another aspect of the present invention relates to a method for the treatment of a disease, the method comprising administering to a patient an effective amount of a composition comprising an antibody of the invention.
  • an antibody of the invention in the manufacture of a medicament for the treatment of a disease.
  • an antibody of the invention for use in the treatment of a disease.
  • compositions comprising an antibody of the invention for the treatment of a disease.
  • a further aspect of the present invention relates to a method for diagnosing a disease, the method comprising
  • an increased amount of the antigen in the sample versus the amount of the antigen in a control sample indicates that the patient is suffering from or is at risk of suffering from the disease.
  • Another aspect of the present invention relates to a method for monitoring the progression of a disease, the method comprising
  • step (iii) repeating steps (i) and (ii) at two or more time intervals, wherein an increase in the amount of the antigen bound to the antibody over time is indicative of a progression of the disease and a reduction in the amount of the antigen bound to the antibody over time is indicative of a regression of the disease.
  • kits for the diagnosis, treatment or monitoring of progression of a disease comprising an antibody of the invention.
  • the disease is selected from a disease characterised by protein misfolding, preferably selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis.
  • the prion diseases are selected from CJD, vCJD, BSE, GSS, FFI and Scrapie.
  • the present invention also relates to a pharmaceutical composition comprising the antibody of the present invention as well as a veterinary composition comprising the antibody of the invention.
  • the route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art.
  • the antibodies and compositions of the invention can be administered to any patient in accordance with standard techniques.
  • the administration can be by any appropriate mode, including parenterally (e.g., intravenous, intramuscular, intraperitoneal, intra- articular, intrathecal), transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter.
  • the dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
  • Administration can be local (e.g., local delivery to the lung by pulmonary administration, e.g., intranasal administration) or systemic as indicated. The skilled clinician will be able to determine the appropriate dosing regimen to treat, suppress or prevent disease.
  • the invention also relates to a drug delivery device comprising the pharmaceutical or veterinary composition of the invention.
  • the drug delivery device can be a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, or rectal delivery device.
  • Examples of such delivery devices include a syringe, a transdermal delivery device (e.g., a patch), a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, and a catheter.
  • a transdermal delivery device e.g., a patch
  • a capsule e.g., a tablet
  • a nebulizer e.g., an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, and a catheter.
  • a transdermal delivery device e.g., a patch
  • a capsule e.
  • the methods and compositions of the invention are for treatment or diagnosis of the disease at an early stage, for example, before symptoms of the disease appear.
  • the methods and compositions of the invention are for treatment or diagnosis of the disease at a clinical stage.
  • the antibodies of the present invention could be used to reverse early neuropathological changes associated with the diseases, for example by removing or neutralising PrPC reverse neurodegeneration. 65 ' 66 .
  • the invention provides compositions (e.g., pharmaceutical and veterinary compositions) comprising an antibody of the invention.
  • the invention also provides a method for treating an individual having a disease or disorder, such as those described herein, comprising administering to said individual a therapeutically effective amount of an antibody of the invention.
  • the disease or disorder is a neurological disease, such as CJD, etc.
  • the invention also provides for use of an antibody of the invention for the manufacture of a medicament for treatment of a disease or disorder.
  • the invention also relates to use of an antibody as described herein for use in therapy, diagnosis or prophylaxis.
  • FIG. 1 shows a schematic representation of use of Dynabeads M-450 to immunoisolate Congo red-bound prion protein.
  • Immunomagnetic isolation of Congo red-bound prion proteins from brain homogenates A schematic outline).
  • a & B After overnight incubation of the sheep anti-mouse IgG coupled 4.5 ⁇ m Dynabeads to anti-PrP mAb, the immunocomplex was added to scrapie-infected brain homogenates (pre-incubated with Congo red) and subsequently incubated for 2 hr at room temperature.
  • C & D The immunocomplex bound to Congo red-bound prion protein was isolated using the MPC-S magnetic device (Dynal), and finally E. This immunocomplex, called PrP/CR-Dynabeads, was used for immunisations of mice and camels;
  • Figure 2 shows adsorption of Congo red bound-PrP Sc is proportional to mAb concentration. Decreasing ten-fold dilutions of mAb (starting at 50 mg/ml) were adsorbed to Dynabeads then 1 x 10 7 of these beads were added to 50ul of a 10% RML brain homogenate bound to Congo red then spiked into an equal volume of 10% Prn-p 0/0 brain homogenate;
  • FIG. 3 shows optimal detection and depletion of Congo red bound-PrP Sc from homogenised brain.
  • Figure 4 shows the effect of medium-term cold storage of the mAb-Dynabeads immunocomplex on depletion of Congo red bound-PrP Sc . mAb-Dynabeads maintain their ability to capture Congo red bound-PrP Sc over time;
  • Figure 5 shows that mAb-Dynabeads bind only a proportion of the Congo red bound-PrP Sc in a 10% dilution.
  • Congo red bound-RML brain homogenate spiked into brain homogenate derived from Prn-p m mice in a 1:10 ratio was proteinase K (PK) treated then incubated with decreasing ten- fold dilutions of mAb-Dynabeads (starting from 1 x 10 7 to 1 x 10 4 ). Both bead- bound protein (b) and supernatant (s) were assayed for PrP Sc levels.
  • Brain homogenate derived from Prn-p m mice (KO) was also included as negative control;
  • Figure 6 shows the effect of Dynabeads concentration on Congo red bound-PrP Sc depletion from various concentrations (1:100, 1:20, and 1/10) of 10% w/v scrapie-infected brain homogenates spiked into Prn-p 00 brain homogenate.
  • 1 x 10 4 to 1 x 10 7 increasing concentration of Dynabeads was incubated with a saturable amount of anti-PrP mAb antibody. Incubation time overnight at 2 - 8°C. Depletion in varying Congo red bound-scrapie brain : Prn-p 010 ratios;
  • Figure 7 shows the effect of incubation time (2 - 8°C) on depletion/binding of Congo red bound-PrP Sc using Dynabeads (1 x 10 7 beads).
  • a 10% Congo red bound-scrapie-infected brain homogenate was incubated with mAb-Dynabreads overnight and samples were analysed at various time points starting from 1 to 24 hours, then amounts of bead-bound and after- wash supernatant PrP Sc were assessed.
  • PrP Sc binding to mAb-Dynabeads is time dependent; and
  • Figure 8 shows treatment of prion-permissive cell lines (ScN2a).
  • Appropriate antibody dilutions of PRIOV and Normal Camel Serum (l ⁇ g and 25 ⁇ g) were added to 1 x 10 4 ScN2a cells with medium in 6 well plates, and left to incubate for twenty-four hours at 37°C (5% CO 2 ). The cells were then removed from the wells by gentle scraping using a rubber syringe, and centrifuged at 800 rpm for 5 minutes.
  • the invention relates to novel antibodies and to methods of their production. Also described are various uses of the novel antibodies.
  • the term "about” means plus or minus 20%, more preferably plus or minus 10%, even more preferably plus or minus 5%, most preferably plus or minus 2%.
  • a "functional derivative, variant or analogue thereof is interpreted to mean an antibody preferably having at least about 50%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, the binding affinity of an antibody as described herein. Such binding affinity can readily be assessed using any suitable method, such as those described herein or known in the art.
  • antibody or antibody fragment refers to an antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such as a Fab, F(ab')2, Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B- cells, hybridomas, transfectomas, yeast or bacteria.
  • an antibody for example IgG, IgM, IgA, IgD or IgE
  • fragment such as a Fab, F(ab')2, Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody
  • analogue as used herein referring to a polypeptide means a modified peptide wherein one or more amino acid residues of the peptide have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the peptide and/or wherein one or more amino acid residues have been deleted from the peptide and or wherein one or more amino acid residues have been added to the peptide.
  • Such addition or deletion of amino acid residues can take place at the N-terminal of the peptide and/or at the C-terminal of the peptide or they can be within the peptide.
  • treatment means treatment of an existing disease and/or prophylactic treatment in order to prevent incidence of a disease.
  • target antigen means an antigen to which the antibodies of the invention are desired to bind.
  • non-target antigen means an antigen to which the antibodies of the invention are desired not to bind.
  • present invention is not limited to antibodies which bind only target antigens and not non-target antigens. In this respect, some antibodies of the invention are able to bind to both target antigens and non-target antigens, whilst other antibodies of the invention are able to bind selectively to target antigens with limited or no binding to non-target antigens.
  • stablising agent means, in particular, an agent that is capable of stablising the conformational structure of a target antigen. Examples include Congo red and Thioflavin T.
  • the term "host” means, for example, a biological system that is capable of producing antibodies against an antigen introduced into that system.
  • examples include an animal host, in particular, rabbits, mice, camelids (Alpacas, Llamas, Camels), rats, sheep and cows.
  • conventional antibody means an antibody which has been previously found to have the desired binding activity.
  • a conventional antibody could be a commercially available antibody or it could, for example, be an antibody previously discovered during the course of previous unpublished experimental work. Put another way, a conventional antibody is a known or previously identified antibody.
  • a conventional antibody could be an antibody identified by the methods described herein. In such an embodiment, once an antibody has been identified by the methods described herein it will become an antibody known to have the desired binding activity.
  • animal includes mammals.
  • mammals includes humans.
  • room temperature means about 21 0 C.
  • the term "specific" means that the antibody has binding specificity for the antigen in question.
  • Congo red is known to bind PrP c and is thought to further stabilize protein conformation 67 . Furthermore, Congo red renders native PrP Sc resistant to denaturation by boiling in SDS. Neither the properties of PrP c nor those of predenatured PrP Sc are changed by the addition of Congo red . In order to study conformational changes induced by Congo red binding to PrP Sc , scrapie infected brain homogenate was first pre-incubated with an appropriate concentration of Congo red with continous rotation for at least 2h.
  • Dynabeads M-450 Sheep anti-Mouse IgG are designed for immunomagnetic separation. Their size is particularly suitable for cell isolation but they are also used for immunoprecipitation of proteins and other purposes. These Dynabeads bind to defined cell surface antigens and other antigens through a mouse primary antibody of the IgG-class. The target cells rosetted with Dynabeads M-450 Sheep anti-Mouse IgG as well as immunoprecipitates can be easily isolated by applying a magnet (Dynal MPC) on the wall of the test tube for 1-2 minutes.
  • a magnet Dynabeads M-450 Sheep anti-Mouse IgG
  • Dynabeads M-450 sheep anti-mouse IgG are monosized, superparamagnetic, polystyrene beads with affinity purified Sheep anti-Mouse IgG covalently bound to the surface. These secondary antibodies bind both heavy and light chain of mouse IgG and IgM light chain. Human cross reactivity is minimal.
  • optimisation of cell yield and purity is, apart from the chosen antibody, especially dependent on two of the variables involved, the number of beads used and the incubation time.
  • the manufacturer recommends the use of 1-2 x 10 7 Dynabeads/ml of sample, and a 10-30 minute incubation time.
  • the affinity and avidity of the primary antibody should be considered as the most important factor in the successful use of Dynabeads M-450 sheep anti-mouse IgG.
  • Concentration of Dynabeads M-450 Sheep anti-Mouse IgG during separation is the single most important parameter for obtaining a good yield, provided that a good primary antibody is used.
  • a concentration of 1- 2 x 10 7 Dynabeads per ml sample is generally sufficient to reach the plateau in the binding kinetics after 20 min incubation.
  • Dynabeads M450 Sheep anti-mouse IgG coupled 4.5 ⁇ m immunomagnetic particles (Dynabeads M450) were used.
  • the Dynabeads M450 were washed 4 times in sterile PBS to remove preservatives, and resuspended with an excess of commercial anti-prion antibody (mAb) to achieve maximal saturation.
  • the Dynabeads/mAb solution was rotated continuously overnight at 4 0 C. After seven washes to remove unbound mAb, the Dynabeads were resuspensed at 10 7 / ml in sterile PBS, and then stored at 4 0 C until use.
  • IxIO 6 mAb-coated Dynabeads were incubated with lOO ⁇ l of 1% scrapie infected brain homogenate bound to Congo red (as determined by SDS- PAGE) for at least 2 hours at room temperature. The final immuno-isolate was washed 7 times and resuspended in PBS. This immunocomplex, called PrP/CR-Dynabeads was subsequently used for immunisations.
  • Anti-PrP/CR-Dynabeads mAbs called PrioC (mouse) and PrioV (camel) mAbs were produced as described above.
  • PrP Sc was first digested with proteinase K (50 ⁇ g/ml) for 30 minutes at 37 0 C to remove PrP c prior to binding to Congo red. The complex PrP Sc /Congo red was then adsorbed to Dynabeads.
  • Mouse Hybridomas Antibody producing cells
  • Positive hybridomas were repeatedly cloned until stable. For camelid antibodies, these were produced using the E coli system.
  • AU PrioC antibodies produced in mice were of the IgM isotype and PrioV were of the IgG isotype.
  • the concentration of the anti-PrP mAb required to saturate the sheep anti-mouse Dynabeads was optimised by varying the binding concentration and then using the mAb-coated beads to immunoprecipitate the same quantity of Congored bound-PrP Sc from murine scrapie brain homogenates (Fig. 2). These experiments indicated that mAb coupling concentrations > 5 x 10 "2 mg/ml saturated the beads. It was also determined that 2.5 x 10 6 PrP/CR-Dynabeads loaded onto each lane gave a readily detectable signal in standard western blots. Optimisation of Congo red bound PrP c from brain homogenate
  • the measure of the relative depletion of Congo red bound-PrP Sc was achieved through direct densitometric analysis of the bead- Congo red bound-PrP Sc after boiling in SDS, rather than substracting it from the post-wash supernatant after the incubation period of the whole irnrnunocomlex as this could give a false measure of the relative amounts present since the concentration of Congo red bound-PrP Sc could exceed the limit and capacity of the beads to adsorb more proteins.
  • Congo red bound-PrP Sc from scrapie-infected brain homogenate spiked into Pm-p m mice brain homogenate was semi-quantitated using the Molecular Dynamics STORM 840 phosphoimager (enhanced chemifluorescent substrate (ECF, Amersham) over a period of 24 hours (Fig. 7). After adding a 10% preparation of the spiked brain homogenate to the mAb- Dynabeads, samples of the immunocomplex were taken at various time points and depletion versus binding was measured with STORM. With the highest concentration of the spiked brain (10%), it was apparent that the incubation period did not achieve complete depletion, but more Congo red bound-PrP Sc was shown to bind to the immunocomplex over time.
  • PrioC mAbs raised against native PrP C /CR-Dynabeads recognises mouse rPrP
  • Clones and subclones of hybridomas derived from spleens of Prn-p 0/0 mice hyperimmunised with PrP s 7CR-Dynabeads were screened for reactivity to alpha or beta recombinant protein.
  • Hybridoma clone supernatants were probed for reactivity with full-length and truncated mouse ⁇ or ⁇ recombinant prion proteins. The majority of these mAb supernatants reacted strongly to both ⁇ and ⁇ conformations of rPrP with ELISA. These supernatants were also screened for total IgG and IgM with isotype ELISA.
  • the isotype response for monoclonal antibodies was predominantly of IgM subtype.
  • PrioC mAbs bind native PrP c/Sc Binding to surface PrP c on U937 cells
  • PrioC mAbs were also probed in order to demonstrate whether binding to surface native PrP c occured. In general PrioC mAbs bound weakly to the surface of U937 cells than either commercial mAb anti-prion, possibly reflecting a lower affinity for PrP .
  • PrP c During its synthesis, PrP c transits through the secretory pathway. The conversion of PrP c to PrP Sc is thought to occur after the former reaches the plasma membrane or is reinternalised for degradation 69 . It was sought to demonstrate cytoplasmic staining of PrP Sc (and PrP c ) in scrapie infected Neuroblastoma cells (N2a) using PrioC mAbs by immunofluorescence labelling. N2a cells were seeded on glass coverslips and grown to 50% confluence for staining of both external and internal PrP. Prior to immunofluorescence staining and imaging, the infected and non infected N2a cells were first lightly paraformaldehyde fixed and permeabilized with Triton X-100.
  • An isotype matched IgM control has shown no binding against PrP, in contrast with the positive control, known to bind PrP with high affinity, that decorated the scrapie-infected N2a cells with a ring around the plasma membrane, indicating strong binding to surface PrP c .
  • PrioC mAbs bound surface PrP on uninfected N2a cells. Since the cells were permeabilized, staining for PrP was also seen inside the cytoplasm, in contrast with mAb positive control where the staining remained at the cell surface forming a ring around it.
  • PrioC mAbs bind native PrP across species
  • Normal and scrapie brain homogenate was proteinase K treated and the PrioC mAbs used to immunoprecipitate PrPC and PrPSc from murine, human, ovine, and bovine brain homogenates.
  • Treatment with proteinase K cleaves approximately 67 amino acids from N- terminus of PrP Sc and completely digests PrP c .
  • Positive control (biotin-conjugate), which detects all glycoforms and fragments of PrP c was used to analyse the immunoprecipitate by western blots after denaturation by boiling in sample buffer for 5 min.
  • PrioC mAbs were used to immunoprecipitate PrP from WT and scrapie-infected brains from mice and human vCJD. An anti-mouse IgM, was used as the negative isotype matched control. It was clear that although PrioC mAbs immunoprecipitated both isoforms of PrP from mouse brain homogenates, they recognised vCJD and not control human brain homogenate.
  • a BSE test evaluated by the EC has previously been developed by the inventors that included the measurement of 48 test positive as well as 152 test negative that were all correctly identified (EU website).
  • the BSE test also included dilution series in order to determine the detection limit of the test, and ranged between 10 "1 and 10 "3 . All 10 "1 , 10 "1 5 and 10 ⁇ 2 scored positive. Only 6/8 and 4/12 scored positive for the lO "2 ' 5 and 10 "3 dilutions respectively.
  • PrioC mAb inhibits propagation of PrP Sc c i.n chronically infected N2a cells
  • a range of concentrations of PrioC and control mAbs were added to ScN2a cultures for 4 days (repeated daily treatment). After day five, 25000, 5000, 1000 ScN2a (the 5000 and 1000 cells were diluted into appropriate concentration of uninfected N2a cells to achieve 25000 total number of cells) cells were placed into an ELISPOT plate in quadruplicate. Levels of PrP Sc in the culture were then analysed with the Scrapie Cell assay. The level of PrP Sc in cells treated with PrioC mAb, compared with the cells treated with isotype matched negative control IgM was dramatically reduced in a dose-dependent manner.
  • PrioC inhibition of PrP Sc level in ScN2a cells was dose-dependent as complete inhibition of PrP Sc was achieved when a titration of the PrioC, ranging from 27 ⁇ g/ml to O.l l ⁇ g/ml was used on the 25000 ScN2a cell dilution.
  • the 5000 ScN2a cell dilution was also efficiently inhibited by the same titration of antibody, but in contrast with the two cell dilutions, the undiluted 1000 ScN2a cells demonstrated less efficiency in the decrease of PrP Sc level, particularly below 1 ⁇ g/ml.
  • Bovine spongiform encephalopathy epidemiological studies. Vet.Rec. 123:638-644. 10.

Abstract

Methods are provided for producing an antibody specific for a target antigen, in particular, antibodies capable of binding to prion proteins. The methods include adding a target antigen stabilising agent to a sample containing a target antigen, allowing the stabilising agent to stabilise the target antigen, incubating the stabilised target antigen with a conventional antibody immobilised on a solid support to form a complex, wherein the immobilised antibody is specific for the target antigen, immunising a host with the complex, and isolating antibodies specific for the target antigen from the host. Also provided are antibodies produced by the methods together with their use in treatment and diagnosis of disease, e.g. prion diseases.

Description

ANTIBODIES SPECIFIC FOR MISFOLDED PROTEINS AND METHODS FOR
THEIR PRODUCTION
BACKGROUND OF THE INVENTION
The present application relates to novel antibodies and to methods for producing antibodies, in particular antibodies capable of binding to prion proteins.
Transmissible spongiform encephalopathies (TSE) or prion diseases comprise a group of invariably fatal neurodegenerative disorders affecting humans and animals, characterised clinically in humans by a rapidly progressive dementia i.e. memory loss and cognitive deficit and neuropathologically, by spongiform degeneration. Prion diseases such as CJD, BSE, together with Alzheimer's disease and certain forms of Parkinsonism are examples of disorders caused by protein misfolding and in prion disease, an abnormally folded protein (called prions or PrP ), derived from a normal cell surface protein (called PrP ), accumulates in the brain and other organs of affected animals. PrP and PrP are identical when denatured, meaning that the toxicity and infectivity of PrPSc is wholly dependent on its conformation, i.e. how it is folded.
The human prion diseases include kuru, Creutzfeldt-Jacob disease (CJD), Gerstmann- Straussler-Scheinker syndrome (GSS), sporadic and familial fatal insomnia (FFI). GSS, FFI and 10-15% of CJD are dominantly inherited disorders associated with specific mutations in the PRNP gene l, whereas 85-90% of CJD occur sporadically, and are not associated with mutations in the human prion protein gene . Iatrogenic CJD has resulted from the use of contaminated neurosurgical instruments, corneal grafts, dura matter grafts, and pituitary derived hormone treatments.
In animals, the prototypic naturally occurring prion disease is scrapie, which affects sheep and goats. Scrapie has been recognised for over 200 years 3 and has a worldwide distribution. Other and more recently recognised animal diseases include transmissible mink encephalopathy, chronic wasting disease of mule, deer, and elk 4, and bovine spongiform encephalopathy (BSE) 5. Prion diseases are characterised by their transmissibility to a wide range of experimental animal species, including mice, hamsters, rats, and demonstrated by the transmission of kuru to chimpanzees 6, and CJD and GSS to primates 7 8. Prion diseases can be transmitted by intracranial inoculation for experimental purposes but the appearance in UK cattle in 1986 of BSE, which rapidly evolved into a major epidemic 9;i0, was widely attributed to transmission of sheep scrapie to cattle via contaminated feed prepared from rendered carcasses 9. The more recently described feline spongiform encephalopathy of domestic cats u and spongiform encephalopathies of a number of zoo animals, the so-called exotic ungulate encephalopathies ' are also recognised as animal prion diseases, and are thought to have resulted from the same contaminated food given to cattle. A need for testing slaughtered cattle over 30 months of age for bovine spongiform encephalopathy has increased the momentum for developing rapid diagnostic tests. However, perhaps the greatest need is for tests with applicability in the sub-clinical phase of prion disease, requiring a specificity that is currently beyond available immunodiagnostic methods.
Both human and animal prion diseases share common histological features, namely spongiform vacuolation, astrocytosis, and neuronal loss. The spongiform change is not seen by the naked eye and vacuoles measure 1 to 50μm in diameter. Proliferation of astrocytic glial cells is another classical marker of prion diseases. This usually accompanies a loss of nerve cells in the surrounding areas of the brain. Amyloid plaques are deposited in the brains of about 15% of prion disease cases and demonstrate the characteristic red/green birefringence when stained with congo red and exposed to polarised light. Initially these plaques were thought to be indistinguishable from those associated with Alzheimer's disease but immunohistochemical work has shown that they are composed of prion deposits. The deposition of amyloid is especially apparent in GSS, Kuru, and vCJD and is a feature of these diseases.
Classical immunisation techniques applied to raise specific anti-prion antibodies have generally been unsuccessful and until recently, only very few antibodies had been produced that recognised any form of PrP l ' . Scrapie-associated fibrils (SAF) were used as immunogen in order to generate an antibody response in both mice and rabbits. Although purified intact infectious SAF did not elicit detectable natural or experimental immune responses 21, a weakly measurable imrmmoreactive antibody reaction was evoked by formic acid-extracted SAF and SDS-solubilised SAF preparations, suggesting that treatment of SAF with detergent or denaturant somehow renders the antigen more immunogenic, perhaps by altering its conformation through a process of denaturation. So-called prion rods were also purified then used to produce anti-PrP antibodies in both rabbits 22 and mice 23. Similar to the SAF preparations, prion rods also undergo substantial and stringent purification steps 22 which could lead to their denaturation and increased immunogenecity while leading to a remarkable reduction in infectivity.
Both the SAF preparations and the prion rods have given rise to weak immune responses but more importantly the antibodies induced bind weakly or not at all to the abnormal isoform of PrPc; PrPSc, and therefore cannot be used as potent immunogens.
Other immunisation approaches have been used, including synthetic peptide vaccination 24"29, nucleic acid-based immunisation 30~32, recombinant PrP 33~35, dimeric PrP 36, and immunisation with scrapie-infected cells 37, none of which has elicited antibody responses specific for PrPSc, except for the 15B3 mAb which apparently binds conformational epitopes on bovine and human PrPSc without recognising the same regions in PrP038.
The peptide vaccination approach has proven to be a useful strategy for generating PrPSc- selective antibodies ' °, although it is proving crucial to further investigate the structure and mechanisms of formation of PrPSc and be able to demonstrate clearly whether these immunogenic motifs are prion disease-associated and not a shared generic epitope with other amyloid-forming diseases .
The nucleic acid-based immunisation strategy 30>32>42 did not prove to be more potent. Here, mice were immunised with DNA 3 or RNA 43 coding for different human prion proteins, following the synthesis of vectors expressing individual genotypes of either the cellular prion gene (PRNP) or mutant forms of the gene.
Full-length recombinant bovine PrP 38 (rbPrP23"231), recombinant murine 4^6 (rmPrP23'231), and recombinant ovine PrP 47 (roPrP23 231) as well as truncated recombinant human PrP 48 (rhPrP91"231) can be expressed in Escherichia coli as an α isoform which can then be folded into β isoforms after reduction of the disulfide bond and lowering the pH to 4.0. The 15B3 49 antibody raised against rbPrP23"231 appears to be selective for the disease-isoform of PrP, although no further application of this antibody has been reported. Monoclonal antibodies to PrP have been successfully raised by various methods in Prn-p0/0 mice 50 that do not express prion protein 23>38-51 53. However, generation of monoclonal antibodies that bind specifically the native conformation PrPSc, has been difficult to produce.
3F4, an IgG2a kappa chain monoclonal antibody 21 was produced by immunising C57BL/6J mice with SAF derived from the hamster 263K prion strain and reacted with hamster but not mouse SAF-derived 263K PrP, indicating strong influence of the host species in determining the antigenic response by the existence of prion epitopes that are species specific.
3F4 was tested with homogenates from normal brain tissues of different species and reacted with both hamster and human PrP in brain preparations but failed to detect PrP in brain tissues from mouse, rat, rabbit, bovine, and sheep. Furthermore, pep-scan analysis of 3F4 has shown that it recognises an epitope present on hamster and human PrP between amino acids 109-113 which is dependent upon the presence of methionines at codon 109 and 112, lacking in other species tested. 3F4 has little or no affinity for native PrPSc.
A report from Schenk et al 54 was the first clear evidence that immunization of transgenic animals prevented the development of beta-amyloid-plaque formation, and reduced the degree of neuropathological progression in an Alzheimer disease model. Following that report, other groups 55;56 have demonstrated the effectiveness of immunisation using amyloid-β peptide in reducing neuropathology in transgenic rodents 57 and humans 58.
Several drugs have been tested for interaction with PrPSc and its accumulation, including Polyanions 59, Iododoxorubicin, Tetracycline 60, Congo red 61, β-sheet breakers 62, Polyene antibiotics 63, Chlorpromazine and Quinacrine 64. Only amphotericine analogue MS8209 has had an appreciable effect on the disease course. Otherwise many of these drugs suppress prion replication in vitro, but fail to show significant efficacy in vivo.
Thus, a need exists for improved methods of producing antibodies that are more effective against antigenic targets such as prion proteins. SUMMARY OF THE INVENTION
According to one aspect of the present invention, there is provided a method for producing an antibody specific for a target antigen, the method comprising:-
(i) adding a target antigen stabilising agent to a sample containing a target antigen;
(ii) allowing the stabilising agent to stabilise the target antigen; (iii) incubating the stabilised target antigen with a conventional antibody immobilised on a solid support to form a complex, wherein the immobilised antibody is specific for the target antigen; (iv) immunising a host with the complex; and (v) isolating antibodies specific for the target antigen from the host.
Preferably, the target antigen is a misfolded protein, more preferably a misfolded prion protein, further preferably PrPSc or PrPc. In some embodiments, the target antigen is misfolded native PrPSo.
Preferably, the stabilising agent is selected from Congo red and Thiofiavin T.
Preferably, step (ii) comprises incubating the stabilising agent with the target antigen for up to about 24 hours, preferably up to about 12 hours, further preferably between about 30 minutes and about 5 hours, more preferably about 1 hour and about 3 hours, most preferably about 1 hour or about 2 hours.
Preferably, step (ii) comprises incubating the stabilising agent with the target antigen with continuous rotation. In preferred embodiments, the stabilising agent is incubated with the target antigen at about room temperature.
Preferably, the stablising agent is present at a concentration of between about 50 μg/ml and about 1000 μg/ml, more preferably between about 100 μg/ml and about 500 μg/ml. hi preferred embodiments, the concentration of stabilising agent is increased over time during step (ii), preferably from about 50 μg/ml to about 1000 μg/ml, most preferably from about 100 μg/ml to about 500 μg/ml. In particularly preferred embodiments, the concentration of stabilising agent is increased continually throughout the incubation time of step (ii). In other embodiments, the concentration of stabilising agent is increased in stepped increments over the incubation time.
Preferably, the sample containing the target antigen is a sample of animal tissue, further preferably spleen or a peripheral blood mononuclear cell (PBMC) or part of the CNS, eg. brain.
Preferably, the animal tissue is homogenised prior to addition of the stabilising agent.
Preferably, the target antigen is treated with a proteinase specific for non-target antigen proteins, prior to step (i), (ii) or (iii). hi preferred embodiments the proteinase is proteinase K.
Preferably the proteinase is present at a concentration of between about 10 μg/ml and about 100 μg/ml, more preferably about 50 μg/ml. Preferably, the target antigen is treated with the proteinase for up to about 2 hours, more preferably for up to about 1 hour, most preferably for about 30 minutes. In some embodiments, the target antigen is treated with the proteinase at a temperature of about 37°C.
Preferably, the animal tissue is from an animal infected with a disease and wherein the target antigen is an indicator or a causative agent of that disease.
Preferably, the disease is selected from a disease characterised by protein misfolding, preferably selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis. Preferably, the prion diseases are selected from CJD, vCJD, BSE, GSS, FFI and Scrapie.
In preferred embodiments, the conventional antibody is immobilised on a solid support by saturating a solid support with the conventional antibody.
hi one embodiment, the conventional antibody is immobilised on the solid support via a second antibody which is immobilised on the solid support and which binds the conventional antibody. Preferably, the host is selected from a mouse, camelid, rabbit, rat, sheep and cow. In preferred embodiments the camelid is selected from an alpaca, camel and llama.
Preferably, prior to immunising the host, the complex is isolated from the sample.
Preferably, the solid support is a bead, further preferably an immunomagnetic bead, more preferably selected from an IgG immunomagnetic bead, IgA immunomagnetic bead, protein G immunomagnetic bead, protein A immunomagnetic bead and streptavidin immunomagnetic bead.
Preferably, the target antigen is immobilised on the solid support by incubating the target antigen with the solid support for up to about 24 hours, preferably for up to about 12 hours, further preferably between about 30 minutes and about 5 hours, more preferably about 1 hour and about 3 hours, most preferably about 1 hour or about 2 hours.
Preferably, the target antigen is incubated with between about 105 and about 107 immunomagnetic particles, more preferably about 106 (25 μl) immunomagnetic particles.
Preferably, the target antigen is incubated with the solid support at between about 00C and about 25°C, preferably between about 20C and about 8°C, more preferably at about 4°C or about room temperature.
According to the invention, there is, therefore, provided a method for producing an anti-PrPSc antibody, the method comprising: -
(i) incubating a scrapie infected brain homogenate with Congo Red;
(ii) immobilising Congo Red bound PrP on an anti-PrP immunomagnetic bead to form a PrP/Congo Red immunomagnetic bead complex (iii) immunising a host with the PrP/Congo Red immunomagnetic bead complex; and (iv) isolating antibodies specific for PrPSc from the host. Preferably, prior to step (i), the scrapie infected brain homogenate is incubated with proteinase K.
Preferably, the scrapie infected brain homogenate is incubated with about 50μg/ml proteinase K at about 37°C for about 30 minutes.
According to another aspect of the present invention, there is provided an antibody produced by a method of the invention.
Preferably, the antibody is a monoclonal antibody. Preferably, the antibody is an antibody fragment. Preferably, the antibody is conjugated to a growth inhibitory agent. Preferably, the antibody is conjugated to a cytotoxic agent, for example a toxin, antibiotic, lytic enzyme or radioactive isotope.
In some embodiments, the antibody is conjugated to a detectable marker, for example a fluorescent marker.
Another aspect of the present invention relates to a method for the treatment of a disease, the method comprising administering to a patient an effective amount of a composition comprising an antibody of the invention.
Also provided is use of an antibody of the invention in the manufacture of a medicament for the treatment of a disease.
Further provided is an antibody of the invention for use in the treatment of a disease.
Another aspect provides a composition comprising an antibody of the invention for the treatment of a disease.
A further aspect of the present invention relates to a method for diagnosing a disease, the method comprising
(i) contacting a sample obtained from a patient at risk of suffering from the disease with an antibody of the invention; (ii) detecting the presence of an antigen in the sample bound to the antibody; wherein the presence of the antigen indicates that the patient is suffering from the disease or is at risk of suffering from the disease.
hi preferred embodiments, an increased amount of the antigen in the sample versus the amount of the antigen in a control sample indicates that the patient is suffering from or is at risk of suffering from the disease.
Another aspect of the present invention relates to a method for monitoring the progression of a disease, the method comprising
(i) contacting a sample obtained from a patient suffering from the disease with an antibody of the invention;
(ii) detecting the presence of an antigen in the sample bound to the antibody; and
(iii) repeating steps (i) and (ii) at two or more time intervals, wherein an increase in the amount of the antigen bound to the antibody over time is indicative of a progression of the disease and a reduction in the amount of the antigen bound to the antibody over time is indicative of a regression of the disease.
Also provided by the present invention is a kit for the diagnosis, treatment or monitoring of progression of a disease, the kit comprising an antibody of the invention.
Preferably, the disease is selected from a disease characterised by protein misfolding, preferably selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis. In preferred embodiments, the prion diseases are selected from CJD, vCJD, BSE, GSS, FFI and Scrapie.
The present invention also relates to a pharmaceutical composition comprising the antibody of the present invention as well as a veterinary composition comprising the antibody of the invention.
The route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art. For therapy, the antibodies and compositions of the invention can be administered to any patient in accordance with standard techniques. The administration can be by any appropriate mode, including parenterally (e.g., intravenous, intramuscular, intraperitoneal, intra- articular, intrathecal), transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter. The dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician. Administration can be local (e.g., local delivery to the lung by pulmonary administration, e.g., intranasal administration) or systemic as indicated. The skilled clinician will be able to determine the appropriate dosing regimen to treat, suppress or prevent disease.
The invention also relates to a drug delivery device comprising the pharmaceutical or veterinary composition of the invention. For example, the drug delivery device can be a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, or rectal delivery device. Examples of such delivery devices, include a syringe, a transdermal delivery device (e.g., a patch), a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, and a catheter.
In preferred embodiments, the methods and compositions of the invention are for treatment or diagnosis of the disease at an early stage, for example, before symptoms of the disease appear.
In some embodiments, the methods and compositions of the invention are for treatment or diagnosis of the disease at a clinical stage. In this respect, the antibodies of the present invention could be used to reverse early neuropathological changes associated with the diseases, for example by removing or neutralising PrPC reverse neurodegeneration. 65'66.
Accordingly, as described above, the invention provides compositions (e.g., pharmaceutical and veterinary compositions) comprising an antibody of the invention. The invention also provides a method for treating an individual having a disease or disorder, such as those described herein, comprising administering to said individual a therapeutically effective amount of an antibody of the invention. In some embodiments, the disease or disorder is a neurological disease, such as CJD, etc. The invention also provides for use of an antibody of the invention for the manufacture of a medicament for treatment of a disease or disorder. The invention also relates to use of an antibody as described herein for use in therapy, diagnosis or prophylaxis.
Example embodiments of the present invention will now be described with reference to the accompanying figures.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a schematic representation of use of Dynabeads M-450 to immunoisolate Congo red-bound prion protein. Immunomagnetic isolation of Congo red-bound prion proteins from brain homogenates (A schematic outline). A & B. After overnight incubation of the sheep anti-mouse IgG coupled 4.5 μm Dynabeads to anti-PrP mAb, the immunocomplex was added to scrapie-infected brain homogenates (pre-incubated with Congo red) and subsequently incubated for 2 hr at room temperature. C & D. The immunocomplex bound to Congo red-bound prion protein was isolated using the MPC-S magnetic device (Dynal), and finally E. This immunocomplex, called PrP/CR-Dynabeads, was used for immunisations of mice and camels;
Figure 2 shows adsorption of Congo red bound-PrPSc is proportional to mAb concentration. Decreasing ten-fold dilutions of mAb (starting at 50 mg/ml) were adsorbed to Dynabeads then 1 x 107 of these beads were added to 50ul of a 10% RML brain homogenate bound to Congo red then spiked into an equal volume of 10% Prn-p0/0 brain homogenate;
Figure 3 shows optimal detection and depletion of Congo red bound-PrPSc from homogenised brain. Congo red bound-RML brain homogenate spiked into brain homogenate derived from Prn-pm mice was proteinase K (PK) treated then incubated with 1 x 107 mAb saturated- Dynabeads (upper panel). In the lower panel, mAb was omitted from the immunocomplex keeping the other conditions identical; Figure 4 shows the effect of medium-term cold storage of the mAb-Dynabeads immunocomplex on depletion of Congo red bound-PrPSc. mAb-Dynabeads maintain their ability to capture Congo red bound-PrPSc over time;
Figure 5 shows that mAb-Dynabeads bind only a proportion of the Congo red bound-PrPSc in a 10% dilution. Congo red bound-RML brain homogenate spiked into brain homogenate derived from Prn-pm mice in a 1:10 ratio was proteinase K (PK) treated then incubated with decreasing ten- fold dilutions of mAb-Dynabeads (starting from 1 x 107 to 1 x 104). Both bead- bound protein (b) and supernatant (s) were assayed for PrPSc levels. Brain homogenate derived from Prn-pm mice (KO) was also included as negative control;
Figure 6 shows the effect of Dynabeads concentration on Congo red bound-PrPSc depletion from various concentrations (1:100, 1:20, and 1/10) of 10% w/v scrapie-infected brain homogenates spiked into Prn-p00 brain homogenate. 1 x 104 to 1 x 107 increasing concentration of Dynabeads was incubated with a saturable amount of anti-PrP mAb antibody. Incubation time overnight at 2 - 8°C. Depletion in varying Congo red bound-scrapie brain : Prn-p010 ratios;
Figure 7 shows the effect of incubation time (2 - 8°C) on depletion/binding of Congo red bound-PrPSc using Dynabeads (1 x 107 beads). A 10% Congo red bound-scrapie-infected brain homogenate was incubated with mAb-Dynabreads overnight and samples were analysed at various time points starting from 1 to 24 hours, then amounts of bead-bound and after- wash supernatant PrPSc were assessed. PrPSc binding to mAb-Dynabeads is time dependent; and
Figure 8 shows treatment of prion-permissive cell lines (ScN2a). Appropriate antibody dilutions of PRIOV and Normal Camel Serum (lμg and 25μg) were added to 1 x 104 ScN2a cells with medium in 6 well plates, and left to incubate for twenty-four hours at 37°C (5% CO2). The cells were then removed from the wells by gentle scraping using a rubber syringe, and centrifuged at 800 rpm for 5 minutes. The pellet was lysed with 50 μl of lysis buffer, then subsequently proteinase K (PK) treated with lug/ml for 25 minutes at 370C in order to digest normal cellular PrP, and finally treated with ABSF to inhibit the PK. The cells were then stored at -20C until further use. Levels of PrPSc in the culture were then analysed by ELISA and Western blot in triplicates for each experiment. Figure 8 shows a clear reduction in PrPSc replication when 10,000 ScN2a cells are treated with 1 and 25 μg of PRIO V. Both concentrations show a significant reduction in prion replication (P<0.001) when compared with the NCS control and the No Ab negative control.
DETAILED DESCRIPTION OF THE INVENTION
The invention relates to novel antibodies and to methods of their production. Also described are various uses of the novel antibodies.
The methods used in the invention and detailed examples of the invention are set out below:
Within this specification embodiments have been described in a way which enables a clear and concise specification to be written, but it is intended and will be appreciated that embodiments may be variously combined or separated without parting from the invention.
Within this specification, the terms "comprises" and "comprising" are interpreted to mean "includes, among other things". These terms are not intended to be construed as "consists of only".
Within this specification, the term "about" means plus or minus 20%, more preferably plus or minus 10%, even more preferably plus or minus 5%, most preferably plus or minus 2%.
Within this specification, a "functional derivative, variant or analogue thereof is interpreted to mean an antibody preferably having at least about 50%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, the binding affinity of an antibody as described herein. Such binding affinity can readily be assessed using any suitable method, such as those described herein or known in the art.
Within this specification, "antibody or antibody fragment" refers to an antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such as a Fab, F(ab')2, Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B- cells, hybridomas, transfectomas, yeast or bacteria.
The term "analogue" as used herein referring to a polypeptide means a modified peptide wherein one or more amino acid residues of the peptide have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the peptide and/or wherein one or more amino acid residues have been deleted from the peptide and or wherein one or more amino acid residues have been added to the peptide. Such addition or deletion of amino acid residues can take place at the N-terminal of the peptide and/or at the C-terminal of the peptide or they can be within the peptide.
Within this specification, the term "treatment" means treatment of an existing disease and/or prophylactic treatment in order to prevent incidence of a disease.
Within this specification, the term "target antigen" means an antigen to which the antibodies of the invention are desired to bind. In the same way "non-target antigen" means an antigen to which the antibodies of the invention are desired not to bind. However, it will be envisaged that the present invention is not limited to antibodies which bind only target antigens and not non-target antigens. In this respect, some antibodies of the invention are able to bind to both target antigens and non-target antigens, whilst other antibodies of the invention are able to bind selectively to target antigens with limited or no binding to non-target antigens.
Within this specification, the term "stablising agent", means, in particular, an agent that is capable of stablising the conformational structure of a target antigen. Examples include Congo red and Thioflavin T.
Within this specification, the term "host" means, for example, a biological system that is capable of producing antibodies against an antigen introduced into that system. Examples include an animal host, in particular, rabbits, mice, camelids (Alpacas, Llamas, Camels), rats, sheep and cows.
Within this specification, the term "conventional antibody" means an antibody which has been previously found to have the desired binding activity. A conventional antibody could be a commercially available antibody or it could, for example, be an antibody previously discovered during the course of previous unpublished experimental work. Put another way, a conventional antibody is a known or previously identified antibody. In some embodiments a conventional antibody could be an antibody identified by the methods described herein. In such an embodiment, once an antibody has been identified by the methods described herein it will become an antibody known to have the desired binding activity.
Within this specification, the term "animal" includes mammals. The term "mammals" includes humans.
Within this specification, the term "room temperature" means about 210C.
Within this specification, the term "specific" means that the antibody has binding specificity for the antigen in question.
EXAMPLES
Preparation of Congo red-bound native prions
In order to increase conformational differences between PrPSc and PrP0, Congo red was used to bind PrPSc. This was then imrnunoadsorbed to Dynabeads and used to immunise camels and mice.
Congo red is known to bind PrP c and is thought to further stabilize protein conformation 67. Furthermore, Congo red renders native PrPSc resistant to denaturation by boiling in SDS. Neither the properties of PrPc nor those of predenatured PrPSc are changed by the addition of Congo red . In order to study conformational changes induced by Congo red binding to PrPSc, scrapie infected brain homogenate was first pre-incubated with an appropriate concentration of Congo red with continous rotation for at least 2h. The mixture was than added to the 35-Dynabeads immunocomplex and incubated for 2h at RT (see preparation of Dynabeads-adsorbed antigen discussed below) Dynabeads® M-450 sheep anti-mouse IgG
Dynabeads M-450 Sheep anti-Mouse IgG are designed for immunomagnetic separation. Their size is particularly suitable for cell isolation but they are also used for immunoprecipitation of proteins and other purposes. These Dynabeads bind to defined cell surface antigens and other antigens through a mouse primary antibody of the IgG-class. The target cells rosetted with Dynabeads M-450 Sheep anti-Mouse IgG as well as immunoprecipitates can be easily isolated by applying a magnet (Dynal MPC) on the wall of the test tube for 1-2 minutes.
Dynabeads M-450 sheep anti-mouse IgG are monosized, superparamagnetic, polystyrene beads with affinity purified Sheep anti-Mouse IgG covalently bound to the surface. These secondary antibodies bind both heavy and light chain of mouse IgG and IgM light chain. Human cross reactivity is minimal.
Using the Dynabeads protocol (Tayebi et al., MoI Med 2004), optimisation of cell yield and purity is, apart from the chosen antibody, especially dependent on two of the variables involved, the number of beads used and the incubation time. The manufacturer recommends the use of 1-2 x 107 Dynabeads/ml of sample, and a 10-30 minute incubation time.
The affinity and avidity of the primary antibody should be considered as the most important factor in the successful use of Dynabeads M-450 sheep anti-mouse IgG. The reactivity to mouse IgG1, IgG23 and IgG2), is very good, while the reactivity to IgG3 is low due to the fact that this subclass is sparse in normal mouse serum. Concentration of Dynabeads M-450 Sheep anti-Mouse IgG during separation is the single most important parameter for obtaining a good yield, provided that a good primary antibody is used. A concentration of 1- 2 x 107 Dynabeads per ml sample is generally sufficient to reach the plateau in the binding kinetics after 20 min incubation.
Preparation of Dynabeads-adsorbed antigen
Sheep anti-mouse IgG coupled 4.5μm immunomagnetic particles (Dynabeads M450) were used. The Dynabeads M450 were washed 4 times in sterile PBS to remove preservatives, and resuspended with an excess of commercial anti-prion antibody (mAb) to achieve maximal saturation. The Dynabeads/mAb solution was rotated continuously overnight at 40C. After seven washes to remove unbound mAb, the Dynabeads were resuspensed at 107 / ml in sterile PBS, and then stored at 40C until use. IxIO6 mAb-coated Dynabeads were incubated with lOOμl of 1% scrapie infected brain homogenate bound to Congo red (as determined by SDS- PAGE) for at least 2 hours at room temperature. The final immuno-isolate was washed 7 times and resuspended in PBS. This immunocomplex, called PrP/CR-Dynabeads was subsequently used for immunisations.
Production of murine (PrioC) and camelid (PrioV) monoclonal antibodies
Anti-PrP/CR-Dynabeads mAbs called PrioC (mouse) and PrioV (camel) mAbs were produced as described above.
Both mice and camels were immunised subcutaneously with PrP/CR-Dynabeads in CFA then in IFA for subsequent boostings. PrPSc was first digested with proteinase K (50μg/ml) for 30 minutes at 370C to remove PrPc prior to binding to Congo red. The complex PrPSc/Congo red was then adsorbed to Dynabeads. Mouse Hybridomas (Antibody producing cells) were screened for reactivity to both native PrPc and PrPSc. Positive hybridomas were repeatedly cloned until stable. For camelid antibodies, these were produced using the E coli system.
AU PrioC antibodies produced in mice were of the IgM isotype and PrioV were of the IgG isotype.
Optimisation of commercial anti-prion antibodies
The concentration of the anti-PrP mAb required to saturate the sheep anti-mouse Dynabeads was optimised by varying the binding concentration and then using the mAb-coated beads to immunoprecipitate the same quantity of Congored bound-PrPSc from murine scrapie brain homogenates (Fig. 2). These experiments indicated that mAb coupling concentrations > 5 x 10"2 mg/ml saturated the beads. It was also determined that 2.5 x 106 PrP/CR-Dynabeads loaded onto each lane gave a readily detectable signal in standard western blots. Optimisation of Congo red bound PrP c from brain homogenate
Increasing amounts of Congo red bound-PrP c were titrated in, in order to achieve maximal specific antigen loading onto Dynabeads saturated with mAb, keeping the concentration of other brain constituents constant by spiking increasing quantities of scrapie brain homogenate into decreasing volumes of homogenates (at equivalent w/v) derived from Prn-p0/0 mice (Fig. 3). Again Congo red bound-PrPSo binding to the bead surface was highly dependent on the presence of the capturing mAb, although at the highest concentrations used, some nonspecific adsorption to the surface of the Dynabeads was evident. When the supernatant from which PrP had been adsorbed to the Dynabeads was assayed for residual PrPSc by western blotting, only partial depletion of the protein was observed under adsorption conditions used, an indication of the inefficacy of PMSF in inactivating the proteinase K.
Effect of medium-term cold storage of the mAb-Dynabeads immunocomplex on depletion of Congo red bound-PrPSc
It was investigated whether medium term storage of the immunocomplex mAb-Dynabeads in the laboratory fridge had any direct and detrimental effect on the adsorption of Congo red bound-PrPSc from the highest (10%) spiked terminally-sick scrapie brain homogenates into PrP -deficient brain homogenate (Fig. 4)
Surprisingly, after storage at 2-80C, there was little change in the detection of Congo red bound-PrPSc after densitometry analysis using Molecular Dynamics STORM 840 phosphoimager (enhanced chemifluorescent substrate (ECF, Amersham) over a period of 60 days.
It was then determined whether the PrPSc-Dynabeads were immunogenic in vivo in camels and mice. Optimising the Dynabead concentration needed to achieve maximal Congo red bound- PrPSc depletion
To determine the concentration of the relative amount of Dynabeads needed to achieve maximal depletion of Congo red bound-PrPSo molecules, a 10% w/v solution of terminally- sick scrapie brain homogenate bound to Congo red was spiked into Prn-pm brain homogenate (1:10 ratio) using decreasing amount of beads (ten-fold) ranging from 107 to 104 and was subsequently immunoprecipitated (Fig. 5 & Fig. 6). Both Congo red bound-PrPSc adsorbed Dynabeads and the supernatants were assayed. The amount of Congo red bound-PrPSc adsorbed to Dynabeads was inversely proportional to the amount detected in the supernatant using mAb. Relative saturation of the Dynabeads was achieved at 1 x 106 beads, then plateaued above this amount. Full depletion of Congo red bound-PrPSc was not achieved as shown by the increasing PrP c signal in the supernatant when the bead concentration was reduced, and a poor signal has been detected with bead-Congo red bound-PrPSc when the beads concentration was below 105.
Using the Molecular Dynamics STORM 840 phosphoimager (enhanced chemifluorescent substrate (ECF, Amersham), a semi-quantitative measure was performed in order to estimate the relative expression levels of Congo red bound-PrPSc depleted from a 10% w/v solutions of terminally-sick scrapie brain homogenates that were first mixed in varying ratios (Scrapie : Prn-pm) and incubated with mAb-Dynabeads, including 1 :100, 1:20, and 1 :10. The measure of the relative depletion of Congo red bound-PrPSc was achieved through direct densitometric analysis of the bead- Congo red bound-PrPSc after boiling in SDS, rather than substracting it from the post-wash supernatant after the incubation period of the whole irnrnunocomlex as this could give a false measure of the relative amounts present since the concentration of Congo red bound-PrPSc could exceed the limit and capacity of the beads to adsorb more proteins.
This has been shown when the highest concentration of the brain homogenate was used (1:10) and compared to other concentrations (1:20 and 1:100) since the same number of beads could deplete more efficiently when lower concentration of the brain homogenate was used. In conclusion, Congo red bound-PrPSc binds significantly only when Dynabeads have been precoated with anti-PrP mAb. Moderate depletion of PrPSc from brain homogenates reflects the limited capacity for adsorption of the antigen to their surface.
Effect of incubation time on depletion of Congo red bound-PrPSc
Congo red bound-PrPSc from scrapie-infected brain homogenate spiked into Pm-pm mice brain homogenate was semi-quantitated using the Molecular Dynamics STORM 840 phosphoimager (enhanced chemifluorescent substrate (ECF, Amersham) over a period of 24 hours (Fig. 7). After adding a 10% preparation of the spiked brain homogenate to the mAb- Dynabeads, samples of the immunocomplex were taken at various time points and depletion versus binding was measured with STORM. With the highest concentration of the spiked brain (10%), it was apparent that the incubation period did not achieve complete depletion, but more Congo red bound-PrPSc was shown to bind to the immunocomplex over time. For instance there was a 50% increase in binding over a 24h period. It was also shown that the amount of Congo red bound-PrPSc detected in the supernatant, after washing the PrP- Dynabeads immunocomplex, was inversely proportional to bead-Congo red bound-PrPSc after SDS boiling.
PrioC mAbs (mouse) raised against native PrP C/CR-Dynabeads recognises mouse rPrP
Clones and subclones of hybridomas derived from spleens of Prn-p0/0 mice hyperimmunised with PrPs7CR-Dynabeads were screened for reactivity to alpha or beta recombinant protein. Hybridoma clone supernatants, were probed for reactivity with full-length and truncated mouse α or β recombinant prion proteins. The majority of these mAb supernatants reacted strongly to both α and β conformations of rPrP with ELISA. These supernatants were also screened for total IgG and IgM with isotype ELISA.
PrioC mAbs Isotyping
The isotype response for monoclonal antibodies was predominantly of IgM subtype. Epitope mapping of PrioC mAbs
Overlapping 20-mer peptides spanning the mouse PrP sequence 90-230 were used. Depending on the way the immunogen was prepared, supernatants bound different PrP regions.
PrioC mAbs bind native PrPc/Sc Binding to surface PrPc on U937 cells
PrioC mAbs were also probed in order to demonstrate whether binding to surface native PrPc occured. In general PrioC mAbs bound weakly to the surface of U937 cells than either commercial mAb anti-prion, possibly reflecting a lower affinity for PrP .
PrioC mAbs binding to membrane and cytoplasmic PrPSc/c with N2a cells
During its synthesis, PrPc transits through the secretory pathway. The conversion of PrPc to PrPSc is thought to occur after the former reaches the plasma membrane or is reinternalised for degradation 69. It was sought to demonstrate cytoplasmic staining of PrPSc (and PrPc) in scrapie infected Neuroblastoma cells (N2a) using PrioC mAbs by immunofluorescence labelling. N2a cells were seeded on glass coverslips and grown to 50% confluence for staining of both external and internal PrP. Prior to immunofluorescence staining and imaging, the infected and non infected N2a cells were first lightly paraformaldehyde fixed and permeabilized with Triton X-100.
An isotype matched IgM control has shown no binding against PrP, in contrast with the positive control, known to bind PrP with high affinity, that decorated the scrapie-infected N2a cells with a ring around the plasma membrane, indicating strong binding to surface PrPc. PrioC mAbs bound surface PrP on uninfected N2a cells. Since the cells were permeabilized, staining for PrP was also seen inside the cytoplasm, in contrast with mAb positive control where the staining remained at the cell surface forming a ring around it.
Similarly, when PrioC mAbs were used for staining permeabilized Sc N2a cells, binding was also seen in the cytoplasm. Staining was also apparent on the membrane of these Sc N2a. In conclusion, staining for both groups of cells did not show considerable differences of binding by these PrioC mAbs.
PrioC mAbs bind native PrP across species
Normal and scrapie brain homogenate was proteinase K treated and the PrioC mAbs used to immunoprecipitate PrPC and PrPSc from murine, human, ovine, and bovine brain homogenates. Treatment with proteinase K cleaves approximately 67 amino acids from N- terminus of PrPSc and completely digests PrPc. Positive control (biotin-conjugate), which detects all glycoforms and fragments of PrPc, was used to analyse the immunoprecipitate by western blots after denaturation by boiling in sample buffer for 5 min.
All PrioC mAbs were used to immunoprecipitate PrP from WT and scrapie-infected brains from mice and human vCJD. An anti-mouse IgM, was used as the negative isotype matched control. It was clear that although PrioC mAbs immunoprecipitated both isoforms of PrP from mouse brain homogenates, they recognised vCJD and not control human brain homogenate.
Diagnostic value of PrioC mAbs
A BSE test evaluated by the EC has previously been developed by the inventors that included the measurement of 48 test positive as well as 152 test negative that were all correctly identified (EU website). The BSE test also included dilution series in order to determine the detection limit of the test, and ranged between 10"1 and 10"3. All 10"1, 10"1 5 and 10~2 scored positive. Only 6/8 and 4/12 scored positive for the lO"2'5 and 10"3 dilutions respectively.
In order to mimic the same format of testing and analysis, PrioC mAb were used. Nine New Zealand control bovine homogenates were also used. Test positive have all scored positive with PrioC mAb (mean reading compared to cut-off). All 10"1 series scored positive and none of the 10~2 as well as 10" dilution series scored positives. See table below for details of the results. Table 1 : Overall results including test positives and negatives as well as dilution series using PrioC mAb
Figure imgf000024_0001
Although the size of the samples was limited and considerably smaller than that used for the BSE EU test, the PrioC mAbs seemed to perform similarly when undiluted samples and dilution series of 10" were used. In contrast none of the 10" and 10" dilution series were detected in this system. In conclusion, the PrioC antibodies raised against native prion proteins have shown promise as diagnostic tools and proved to be very effective in detecting concentrations of PrPSo molecules in the 10"1 homogenates.
PrioC mAb inhibits propagation of PrP Scc i.n chronically infected N2a cells
A range of concentrations of PrioC and control mAbs were added to ScN2a cultures for 4 days (repeated daily treatment). After day five, 25000, 5000, 1000 ScN2a (the 5000 and 1000 cells were diluted into appropriate concentration of uninfected N2a cells to achieve 25000 total number of cells) cells were placed into an ELISPOT plate in quadruplicate. Levels of PrPSc in the culture were then analysed with the Scrapie Cell assay. The level of PrPSc in cells treated with PrioC mAb, compared with the cells treated with isotype matched negative control IgM was dramatically reduced in a dose-dependent manner.
In contrast, 4-day treatment with control, did not reduce the level of PrPSc in ScN2a cultures even at high concentrations.
PrioC inhibition of PrPSc level in ScN2a cells was dose-dependent as complete inhibition of PrPSc was achieved when a titration of the PrioC, ranging from 27μg/ml to O.l lμg/ml was used on the 25000 ScN2a cell dilution. The 5000 ScN2a cell dilution was also efficiently inhibited by the same titration of antibody, but in contrast with the two cell dilutions, the undiluted 1000 ScN2a cells demonstrated less efficiency in the decrease of PrPSc level, particularly below 1 μg/ml.
It should be understood that various changes and modifications to the presently preferred embodiments described herein will be apparent to those skilled in the art. Such changes and modifications can be made without departing from the spirit and scope of the present invention and without diminishing its attendant advantages. It is therefore intended that such changes and modifications are covered by the appended claims.
Reference List
1. LaplancheJ.L. 1994. [Molecular basis of familial and sporadic forms of human prion diseases]. Transfus.Clin.Biol. 1:345-353.
2. Prusiner,S.B. and K.K.Hsiao. 1994. Human prion diseases. Ann.Neurol 35:385-395.
3. McGowan,J.P. 1922. Scrapie in sheep. Scott.J.Agric. 5:365-375.
4. Williams,E.S. and S. Young. 1980. Chronic wasting disease of captive mule deer: a spongiform encephalopathy. J Wildl.Dis. 16:89-98.
5. Wells,G.A.H., A.C.Scott, C.T.Johnson, R.F.Gunning, R.D.Hancock, M.Jeffrey, M.Dawson, and R.Bradley. 1987. A novel progressive spongiform encephalopathy in cattle. Vet.Rec. Oct 31 :419-420.
6. Gajdusek,D.C, C.J.Jr.Gibbs, and M.P.Alpers. 1966. Experimental transmission of a kuru-like syndrome to chimpanzees. Nature 209:794-796.
7. LampertJP.W., D.C.Gajdusek, and C.J.Gibbs, Jr. 1972. Subacute spongiform virus encephalopathies. Scrapie, Kuru and Creutzfeldt- Jakob disease: a review. Am.J.Pathol. 68:626-652.
8. Gibbs,C.J., Jr. and D.C.Gajdusek. 1972. Transmission of scrapie to the cynomolgus monkey (Macaca fascicularis). Nature 236:73-74.
9. WilesmithJ.W., G.A.Weils, M.P.Cranwell, and J.B.Ryan. 1988. Bovine spongiform encephalopathy: epidemiological studies. Vet.Rec. 123:638-644. 10. Anderson,R.M., C.A.Donnelly, N.M.Ferguson, M.E.J .Woolhouse, C.J.Watt, HJ.Udy, S.MaWhinney, S.P.Dunstan, T.R.E.Southwood, J.W.Wilesmith, J.B.M.Ryan, L.J.Hoinville, J.E.Hillerton, A.R.Austin, and G.A.H.Wells. 1996. Transmission dynamics and epidemiology of BSE in British cattle. Nature 382:779-788.
11. Wyatt,J.M., G.R.Pearson, T.N.Smerdon, T.J.Gruffydd-Jones, G.A.H.Wells, and J.W.Wilesmith. 1991. Naturally occurring scrapie-like spongiform encephalopathy in five domestic cats. Vet.Rec. 129:233-236.
12. Jeffrey,M. and G.A.Wells. 1988. Spongiform encephalopathy in a nyala (Tragelaphus angasi). Veterinary Pathology 25:398-399.
13. Kirkwood,J.K., G.A.Wells, J.W.Wilesmith, A.A.Cunningham, and S.I.Jackson. 1990. Spongiform encephalopathy in an arabian oryx (Oryx leucoryx) and a greater kudu (Tragelaphus strepsiceros) [see comments]. VetRec. 127:418-420.
14. Kascsak,R.J., R.Rubenstein, P.A.Merz, M.Tonna-DeMasi, R.Fersko, R.I.Carp, H.M.Wisniewski, and H.Diringer. 1987. Mouse polyclonal and monoclonal antibody to scrapie-associated fibril proteins. J. Virol. 61:3688-3693.
15. Bendheim,P.E., R.A.Barry, S.J.DeArmond, D.P.Stites, and S.B.Prusiner. 1984. Antibodies to a scrapie prion protein. Nature 310:418-421.
16. Bode,L., M.Pocchiari, H.Gelderblom, and H.Diringer. 1985. Characterization of antisera against scrapie-associated fibrils (SAF) from affected hamster and cross-reactivity with SAF from scrapie-affected mice and from patients with Creutzfeldt-Jakob disease. J Gen. Virol. 66:2471-2478. 17. Kascsak,R.J., R.Rubenstein, P.A.Merz, R.I.Carp, N.K.Robakis, H.M.Wisniewski, and H.Diringer. 1986. Immunological comparison of scrapie-associated fibrils isolated from animals infected with four different scrapie strains. J Virol. 59:676-683.
18. Takahashi,K., M.Shinagawa, S.Doi, S.Sasaki, H.Goto, and G.Sato. 1986. Purification of scrapie agent from infected animal brains and raising of antibodies to the purified fraction. Microbiology and Immunology 30:123-131.
19. Demart,S., J.G.Fournier, C.Creminon, Y.Frobert, F.Lamoury, D.Marce, C.Lasmezas, D.Dormont, J.Grassi, and J.P.Deslys. 1999. New insight into abnormal prion protein using monoclonal antibodies. Biochemical and Biophysical Research Communications 265:652-657.
20. Rubenstein,R., RJ.Kascsak, M.Papini, R.Kascsak, R.I.Carp, G.LaFauci, R.Meloen, and J.Langeveld. 1999. Immune surveillance and antigen conformation determines humoral immune response to the prion protein immunogen. Journal ofNeurovirology 5:401-413.
21. Kascsak,R.J., R.Rubenstein, P.A.Merz, M.Tonna DeMasi, R.Fersko, R.I.Carp, H.M.Wisniewski, and H.Diringer. 1987. Mouse polyclonal and monoclonal antibody to scrapie-associated fibril proteins. J Virol. 61:3688-3693.
22. Prusiner,S.B., D.Groth, A.Serban, R.Koehler, D.Foster, M.Torchia, D.Burton, S.L.Yang, and SJ.DeArmond. 1993. Ablation of the prion protein (PrP) gene in mice prevents scrapie and facilitates production of anti-PrP antibodies. Proc Natl Acad Sd USA 90:10608-10612.
23. Williamson,R.A., D.Peretz, N.Smorodinsky, R.Bastidas, H.Serban, I.Mehlhorn, SJ.DeArmond, S.B.Prusiner, and D.R.Burton. 1996. Circumventing tolerance to generate autologous monoclonal antibodies to the prion protein. Proceedings of the National Academy of Sciences of the United States of America 93:7279-7282.
24. Harmeyer,S., E.Pfaff, and M.H.Groschup. 1998. Synthetic peptide vaccines yield monoclonal antibodies to cellular and pathological prion proteins of ruminants. Journal of General Virology 79:937-945.
25. Matsushita,K., H.Horiuchi, S.Furusawa, M.Horiuchi, M.Shinagawa, and H.Matsuda. 1998. Chicken monoclonal antibodies against synthetic bovine prion protein peptide. J Vet.Med Sci 60:777-779.
26. Demart,S., J.G.Fournier, C.Creminon, Y.Frobert, F.Lamoury, D.Marce, C.Lasmezas, D.Dormont, J.Grassi, and J.P.Deslys. 1999. New insight into abnormal prion protein using monoclonal antibodies. Biochemical and Biophysical Research Communications 265:652-657.
27. Takahashi,H., R.H.Takahashi, H.Hasegawa, M.Horiuchi, M.Shinagawa, T.Yokoyama, K.Kimura, M.Haritani, T.Kurata, and K.Nagashima. 1999. Characterization of antibodies raised against bovine-PrP -peptides. Journal ofNeurovirology 5:300-307.
28. Hanan,E., S.A.Priola, and B.Solomon. 2001. Antiaggregating antibody raised against human PrP 106-126 recognizes pathological and normal isoforms of the whole prion protein. Cell MoI. Neurobiol. 21:693-703.
29. Souan,L., R.Margalit, O.Brenner, I.R.Cohen, and F.Mor. 2001. Self prion protein peptides are immunogenic in Lewis rats. J.Autoimmun. 17:303-310. 30. Krasemann,S., M.Groschup, G.Hunsmann, and W.Bodemer. 1996. Induction of antibodies against human prion proteins (PrP) by DNA-mediated immunization of PrP0/0 mice. J.Immunol.Methods 199: 109- 118.
31. Krasemann,S., T.Jύrgens, and W.Bodemer. 1999. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. Journal of Biotechnology 73:119-129.
32. Krasemann,S., W.Bodemer, and M.H.Groschup. 1998. Generation of monoclonal antibodies against prion proteins by nucleic-acid-mediated immunization of PrP mice. Vaccines 97:265-272.
33. Koller,M.F., T.Grau, and P. Christen. 2002. Induction of antibodies against murine full- length prion protein in wild-type mice. J.Neuroimmunol. 132:113-116.
34. BetempSjD. and T.Baron. 2001. Molecular specificities of antibodies against ovine and murine recombinant prion proteins. Biochemical and Biophysical Research Communications 281:101-108.
35. Zanusso,G., D.Liu, S.Ferrari, I.Hegyi, X.Yin, A.Aguzzi, S.Hornemann, S.Liemann, R.Glockshuber, J.C.Manson, P.Brown, R.B.Petersen, P.Gambetti, and M.S.Sy. 1998. Prion protein expression in different species: analysis with a panel of new mAbs. Proc Natl Acad Sd U.S.A. 95:8812-8816.
36. Gilch,S., F.Wopfner, I.Renner-Muller, E.Kremmer, C.Bauer, E.Wolf, G.Brem, M.H.Groschup, and H.M.Schatzl. 2003. Polyclonal anti-PrP auto-antibodies induced with dimeric PrP interfere efficiently with PrPSc propagation in prion-infected cells. J Biol.Chem. 37. Nakamura,N., K.Miyamoto, M.Shimokawa, N.Nishida, S.Mohri, T.Kitamoto, H.Horiuchi, S.Furusawa, and H.Matsuda. 2003. Generation of antibodies against prion protein by scrapie-infected cell immunization of PrP(O/O) mice. Hybrid.Hybridomics. 22:263-266.
38. Korth,C, B.Stierli, P.Streit, M.Moser, O.Schaller, R.Fischer, W.Schulz-Schaeffer, H.Kretzschmar, A.Raeber, U.Braun, F.Ehrensperger, S.Hornemann, R.Glockshuber, R.Riek, M.Billeter, K.Wϋthrich, and B.Oesch. 1997. Prion (PrPSc)-specific epitope defined by a monoclonal antibody. Nature 390:74-77.
39. Curin,e., V, M.Bresjanac, M.Popovic, H.K.Pretnar, V.Galvani, R.Rupreht, M.Cernilec, T.Vranac, I.Hafher, and R.Jerala. 2003. Monoclonal antibody against a peptide of human prion protein discriminates between Creutzfeldt-Jacob's disease- affected and normal brain tissue. J Biol.Chem.
40. Paramithiotis,E., M.Pinard, T.Lawton, S.LaBoissiere, V.L.Leathers, W.Q.Zou, L.A.Estey, J.Lamontagne, M.T.Lehto, L.H.Kondejewski, G.P.Francoeur, M.Papadopoulos, A.Haghighat, S.J.Spatz, M.Head, R.Will, J.Ironside, K.O'Rourke, Q.Tonelli, H.C.Ledebur, A.Chakrabartty, and N.R.Cashman. 2003. A prion protein epitope selective for the pathologically misfolded conformation. Nat Med.
41. O'Nuallain,B. and R.Wetzel. 2002. Conformational Abs recognizing a generic amyloid fibril epitope. Proc.Natl.Acad.Sci U.S.A 99:1485-1490.
42. Krasemann,S., T.Jϋrgens, and W.Bodemer. 1999. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. Journal of Biotechnology 73:119-129. 43. Krasemann,S., T.Jύrgens, and W.Bodemer. 1999. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. Journal of Biotechnology 73:119-129.
44. Hornemann,S., C.Korth, B.Oesch, R.Riek, G.Wider, K.Wϋthrich, and R.Glockshuber. 1997. Recombinant full-length murine prion protein, mPrP(23- 231): purification and spectroscopic characterization. FEBS Lett. 413:277-281.
45. Betemps,D. and T.Baron. 2001. Molecular specificities of antibodies against ovine and murine recombinant prion proteins. Biochemical and Biophysical Research Communications 281:101-108.
46. Liemann,S. and R.Glockshuber. 1999. Influence of amino acid substitutions related to inherited human prion diseases on the thermodynamic stability of the cellular prion protein. Biochemistry 38:3258-3267.
47. Betemps,D. and T.Baron. 2001. Molecular specificities of antibodies against ovine and murine recombinant prion proteins. Biochemical and Biophysical Research Communications 281:101-108.
48. Jackson,G.S., L.L.P.Hosszu, A.Power, A.F.Hill, J.Kenney, H.Saibil, C.J.Craven, J.P.Waltho, A.R.Clarke, and J.Collinge. 1999. Reversible Conversion of Monomelic Human Prion Protein Between Native and Fibrilogenic Conformations. Science 283:1935-1937.
49. Korth,C, K.Kaneko, D.Groth, N.Heye, G.Telling, J.Mastrianni, P.Parchi, P.Gambetti, R. Will, J.Ironside, C.Heinrich, P.Tremblay, S.J.DeArmond, and S.B.Prusiner. 2003. Abbreviated incubation times for human prions in mice expressing a chimeric mouse- human prion protein transgene. Proc.Natl.Acad.Sci U.S.A.
50. Bueler,H., A.Aguzzi, A.Sailer, R.A.Greiner, P.Autenried, M.Aguet, and C.Weissmann. 1993. Mice devoid of PrP are resistant to scrapie. Cell 73:1339-1347.
51. Zanusso,G., D.Liu, S.Ferrari, I.Hegyi, X.Yin, A.Aguzzi, S.Hornemann, S.Liemann, R.Glockshuber, J.C.Manson, P.Brown, R.B.Petersen, P.Gambetti, and M.S.Sy. 1998. Prion protein expression in different species: analysis with a panel of new mAbs. Proc Natl Acad Sd U.S.A. 95:8812-8816.
52. Demart,S., J.G.Fournier, C.Creminon, Y.Frobert, F.Lamoury, D.Marce, C.Lasmezas, D.Dormont, J.Grassi, and J.P.Deslys. 1999. New insight into abnormal prion protein using monoclonal antibodies. Biochemical and Biophysical Research Communications 265:652-657.
53. Yokoyama,T., K.M.Kimura, Y.Ushiki, S.Yamada, A.Morooka, T.Nakashiba, T.Sassa, and S.Itohara. 2001. In vivo conversion of cellular prion protein to pathogenic isoforms, as monitored by conformation-specific antibodies. Journal of Biological Chemistry 276:11265-11271.
54. Schenk,D., R.Barbour, W.Dunn, G.Gordon, H.Grajeda, T.Guido, K.Hu, J.Huang, KJohnson-Wood, K.Khan, D.Kholodenko, M.Lee, Z.Liao, I.Lieberburg, R.Motter, L.Mutter, F.Soriano, G.Shopp, N.Vasquez, C.Vandevert, S.Walker, M.Wogulis, T.Yednock, D. Games, and P.Seubert. 1999. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400:173-177. 55. McLaurin,J., R.Cecal, M.E.Kierstead, X.Tian, A.L.Phinney, M.Manea, J.E.French, M.H.Lambermon, A.A.Darabie, M.E.Brown, C.Janus, M.A.Chishti, P.Horne, D.Westaway, P.E.Fraser, H.T.Mount, M.Przybylski, and P.George-Hyslop. 2002. Therapeutically effective antibodies against amyloid-beta peptide target amyloid-beta residues 4-10 and inhibit cytotoxicity and fibrillogenesis. Nat.Med. 8:1263-1269.
56. Hock,C, U.Konietzko, A.Papassotiropoulos, A.Wollmer, J.Streffer, R.C.von Rotz, G.Davey, E.Moritz, and R.M.Nitsch. 2002. Generation of antibodies specific for beta- amyloid by vaccination of patients with Alzheimer disease. Nat.Med. 8:1270-1275.
57. McLaurinJ., R.Cecal, M.E.Kierstead, X.Tian, A.L.Phinney, M.Manea, J.E.French, M.H.Lambermon, A.A.Darabie, M.E.Brown, C.Janus, M.A.Chishti, P.Horne, D.Westaway, P.E.Fraser, H.T.Mount, M.Przybylski, and P.George-Hyslop. 2002. Therapeutically effective antibodies against amyloid-beta peptide target amyloid-beta residues 4-10 and inhibit cytotoxicity and fibrillogenesis. Nat.Med. 8:1263-1269.
58. Hock,C, U.Konietzko, A.Papassotiropoulos, A.Wollmer, J.Streffer, R.C.von Rotz, G.Davey, E.Moritz, and R.M.Nitsch. 2002. Generation of antibodies specific for beta- amyloid by vaccination of patients with Alzheimer disease. Nat.Med. 8:1270-1275.
59. Ehlers,B. and H.Diringer. 1984. Dextran sulphate 500 delays and prevents mouse scrapie by impairment of agent replication in spleen. J Gen. Virol. 65:1325-1330.
«
60. Forloni,G., S.Iussich, T.Awan, L.Colombo, N.Angeretti, L.Girola, I.Bertani, G.Poli, M.Caramelli, B.M.Grazia, L.Farina, L.Limido, G.Rossi, G.Giaccone, J.W.Ironside, O.Bugiani, M.Salmona, and F.Tagliavini. 2002. Tetracyclines affect prion infectivity. Proc.Natl.Acad.Sci. U.S.A 99: 10849-10854. 61. Caughey,B., D.Ernst, and R.E.Race. 1993. Congo red inhibition of scrapie agent replication. J Virol. 67:6270-6272.
62. Soto,C, RJ.Kascsack, G.P.Saborio, P.Aucouturier, T.Wisniewski, F.Prelli, R.Kascsak, E.Mendez, D.A.Harris, J.Ironside, F.Tagliavini, R.I. Carp, and B.Frangione. 2000. Reversion of prion protein conformational changes by synthetic beta-sheet breaker peptides. Lancet 355:192-197.
63. Adjou,K.T., N.Privat, S.Demart, J.P.Deslys, M.Seman, J.J.Hauw, and D.Dormont. 2000. MS-8209, an amphotericin B analogue, delays the appearance of spongiosis, astrogliosis and PrPres accumulation in the brain of scrapie-infected hamsters. Journal of Comparative Pathology 122:3-8.
64. Korth,C, B.C.May, F.E.Cohen, and S.B.Prusiner. 2001. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc Natl Acad Sci U.S. A 98:9836-9841.
65. Mallucci,G.R., S.Ratte, E.A.Asante, J.Linehan, I.Gowland, J.G.Jefferys, and J.Collinge. 2002. Post-natal knockout of prion protein alters hippocampal CAl properties, but does not result in neurodegeneration. EMBO J. 21 :202-210.
66. Mallucci,G., A.Dickinson, J.Linehan, P.C.Klohn, S.Brandner, and J.Collinge. 2003. Depleting neuronal PrP in prion infection prevents disease and reverses spongiosis. Science 302:871-874.
67. Caspi,S., M.Halimi, A.Yanai, S.B.Sasson, A.Taraboulos, and R.Gabizon. 1998. The anti-prion activity of Congo red. Putative mechanism. J Biol Chem. 273:3484-3489. 68. Casρi,S., M.Halimi, A.Yanai, S.B.Sasson, A.Taraboulos, and R.Gabizon. 1998. The anti-prion activity of Congo red. Putative mechanism. J Biol Chem. 273:3484-3489.
69. BorcheltjD.R., A.Taraboulos, and S.B.Prusiner. 1992. Evidence for synthesis of scrapie prion proteins in the endocytic pathway. J Biol. Chem. 267:16188-16199.

Claims

1. A method for producing an antibody specific for a target antigen, the method comprising: -
(i) adding a target antigen stabilising agent to a sample containing a target antigen;
(ii) allowing the stabilising agent to stabilise the target antigen;
(iii) incubating the stabilised target antigen with a conventional antibody immobilised on a solid support to form a complex, wherein the immobilised antibody is specific for the target antigen;
(iv) immunising a host with the complex; and
(v) isolating antibodies specific for the target antigen from the host.
2. A method according to claim 1 , wherein the target antigen is a misfolded protein.
3. A method according to claim 1 or 2, wherein the target antigen is a prion protein.
4. A method according to claim 3, wherein the prion protein is PrP or PrP .
5. A method according to any preceding claim, wherein the stabilising agent is selected from Congo red and Thioflavin T.
6. A method according to any preceding claim, wherein the sample containing the target antigen is a sample of animal tissue.
7. A method according to claim 6, wherein the animal tissue is selected from spleen, a peripheral blood mononuclear cell (PBMC) and a part of the Central Nervous System (CNS).
8. A method according to claim 6 or 7, wherein the animal tissue is homogenised prior to addition of the stabilising agent.
9. A method according to any of claims 6 to 8, wherein the animal tissue is treated with a proteinase specific for non-target antigen proteins.
10. A method according to any of claims 6 to 9, wherein the animal tissue is from an animal infected with a disease and wherein the target antigen is an indicator of that disease.
11. A method according to claim 10, wherein the disease is characterised by protein misfolding.
12. A method according to claim 10 or 11, wherein the disease is selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis.
13. A method according to any preceding claim, wherein the host is selected from a mouse, camelid, rabbit, rat, sheep and cow.
14. A method according to any preceding claim, wherein prior to immunising the host, the complex is isolated from the sample.
15. A method according to any preceding claim, wherein the conventional antibody is immobilised on the solid support via a second antibody which is immobilised on the solid support and which binds the conventional antibody.
16. A method according to any preceding claim, wherein the solid support is a bead.
17. A method according to any preceding claim, wherein the solid support is an immunomagnetic bead.
18. A method for producing an anti-PrPSc antibody, the method comprising:- (i) incubating a scrapie infected brain homogenate with Congo Red;
(ii) immobilising Congo Red bound PrP on an anti-PrP immunomagnetic bead to form a PrP/Congo Red immunomagnetic bead complex;
(iii) immunising a host with the PrP/Congo Red immunomagentic bead complex; and
(iv) isolating antibodies specific for PrPSc from the host.
19. A method according to claim 18, wherein prior to stem (i), the scrapie infected brain homogenate is incubated with proteinase K.
20. An antibody produced by a method according to any preceding claim.
21. An antibody according to claim 20, wherein the antibody is a monoclonal antibody.
22. An antibody according to claim 20 or 21, wherein the antibody is an antibody fragment.
23. An antibody according to any of claims 20 to 22, wherein the antibody is conjugated to a growth inhibitory agent.
24. An antibody according to any of claims 20 to 23, wherein the antibody is conjugated to a cytotoxic agent, for example a toxin, antibiotic, lytic enzyme or radioactive isotope.
25. A method for the treatment of a disease, the method comprising administering to a patient an effective amount of a composition comprising an antibody acccording to any of claims 20 to 24.
26. Use of an antibody according to any of claims 20 to 24 in the manufacture of a medicament for the treatment of a disease.
27. An antibody according to any of claims 20 to 24 for use in the treatment of a disease.
28. A composition comprising an antibody according to any of claims 20 to 24 for the treatment of a disease.
29. A method for diagnosing a disease, the method comprising:-
(i) contacting a sample obtained from a patient at risk of suffering from the disease with an antibody according to any of claims 20 to 24;
(ii) detecting the presence of an antigen in the sample bound to the antibody; wherein the presence of the antigen indicates that the patient is suffering from the disease or is at risk of suffering from the disease.
30. A method according to claim 29, wherein an increased amount of the antigen in the sample versus the amount of the antigen in a control sample indicates that the patient is suffering from or is at risk of suffering from the disease.
31. A method for monitoring the progression of a disease, the method comprising: -
(i) contacting a sample obtained from a patient suffering from the disease with an antibody according to any of claims 20 to 24;
(ii) detecting the presence of an antigen in the sample bound to the antibody; and
(iii) repeating steps (i) and (ii) at two or more time intervals, wherein an increase in the amount of the antigen bound to the antibody over time is indicative of a progression of the disease and a reduction in the amount of the antigen bound to the antibody over time is indicative of a regression of the disease.
32. A kit for the diagnosis of a disease, the kit comprising an antibody according to any of claims 20 to 24.
33. A method, use, antibody, composition or kit according to any of claims 25 to 32, wherein the disease is characterised by protein misfolding.
34. A method, use, antibody, composition of kit according to any of claims 25 to 33, wherein the disease is selected from prion diseases, Alzeihmer's Disease, Parkinson's Diseases and Diabetes amyloidosis.
35. A pharmaceutical composition comprising an antibody of any of claims 20 to 24.
36. A veterinary composition comprising an antibody of any of claims 20 to 24.
PCT/GB2009/001944 2008-08-08 2009-08-10 Antibodies specific for misfolded proteins and methods for their production WO2010015834A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0814586.4A GB0814586D0 (en) 2008-08-08 2008-08-08 Novel antibodies and methods for their production
GB0814586.4 2008-08-08

Publications (1)

Publication Number Publication Date
WO2010015834A1 true WO2010015834A1 (en) 2010-02-11

Family

ID=39790542

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2009/001944 WO2010015834A1 (en) 2008-08-08 2009-08-10 Antibodies specific for misfolded proteins and methods for their production

Country Status (2)

Country Link
GB (1) GB0814586D0 (en)
WO (1) WO2010015834A1 (en)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
B. PIEKARSKA ET AL.: "Complexation of supramolecular dye Congo red with immunoglobulins. The possible mechanism of dye-induced stabilization of antigen-antibody complexes.", FEBS JOURNAL, vol. 272, no. suppl.1, July 2005 (2005-07-01), pages 74, XP002551502 *
G. ROSSI ET AL.: "Therapeutic approaches to prion diseases.", CLINICS IN LABORATORY MEDICINE, vol. 23, no. 1, 1 March 2003 (2003-03-01), London, GB, pages 187 - 208, XP008070307 *
M. TAYEBI ET AL.: "Disease-associated prion protein elicits immunoglobulin M responses in vivo.", MOLECULAR MEDICINE, vol. 10, no. 7-12, July 2004 (2004-07-01), Cambridge, USA, pages 104 - 111, XP002551501 *
S. CASPI ET AL.: "The anti-prion activity of Congo red. Putative mechanism.", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 273, no. 6, 6 February 1998 (1998-02-06), Birmingham, USA, pages 3484 - 3489, XP002995536 *

Also Published As

Publication number Publication date
GB0814586D0 (en) 2008-09-17

Similar Documents

Publication Publication Date Title
EP1910829B1 (en) Prevention and treatment of synucleinopathic and amyloidogenic disease
DK2361638T3 (en) Beta-1-42-specific monoclonal antibodies with therapeutic properties
US8795664B2 (en) Monoclonal antibodies targeting amyloid beta oligomers
Demart et al. New insight into abnormal prion protein using monoclonal antibodies
US9534044B2 (en) Alpha-synuclein antibodies and uses thereof
Neff et al. Immunotherapy and naturally occurring autoantibodies in neurodegenerative disorders
JP2019218362A (en) N-TERMINAL TRUNCATED AMYLOID β PROTOFIBRILS/OLIGOMERS FOR USE IN TREATING OR DIAGNOSING METHODS FOR ALZHEIMER&#39;S DISEASE
Mably et al. Anti-Aβ antibodies incapable of reducing cerebral Aβ oligomers fail to attenuate spatial reference memory deficits in J20 mice
US11919947B2 (en) Antibody binding active α-synuclein
EP0931551B1 (en) Method of producing therapeutics for transmissible spongiform encephalopathy and non-infective blood and tissue derived products as well as methods of producing these
US20080057057A1 (en) Anti-Lipid Rafts Antibodies
WO2016053610A1 (en) Antibodies against pathological forms of tdp-43 and uses thereof
Arbel et al. Generation of antibodies against prion protein in wild-type mice via helix 1 peptide immunization
Horiuchi et al. Generation of monoclonal antibody that distinguishes PrPSc from PrPC and neutralizes prion infectivity
Miyamoto et al. Inhibition of prion propagation in scrapie-infected mouse neuroblastoma cell lines using mouse monoclonal antibodies against prion protein
WO2010015834A1 (en) Antibodies specific for misfolded proteins and methods for their production
US9796778B1 (en) Antibodies against pathological forms of TDP-43 and uses thereof
Alexandrenne et al. Curative properties of antibodies against prion protein: a comparative in vitro study of monovalent fragments and divalent antibodies
US7550144B2 (en) Prion inhibition
Koo et al. Immunohistochemical detection of prion protein (PrP-Sc) and epidemiological study of BSE in Korea
Miyamoto et al. Chicken antibody against a restrictive epitope of prion protein distinguishes normal and abnormal prion proteins
US20230365665A1 (en) ANTIBODY COMPOSITIONS TARGETING NON-PHOSPHORYLATED a-SYNUCLEIN AGGREGATES
Xiao et al. Preparation of monoclonal antibodies against prion proteins with full-length hamster prp
US20190275125A1 (en) Abeta variants, assay, method and treatment of alzheimer&#39;s disease
JP2004535387A (en) Antibodies that eliminate prion growth and promote infectious clearance

Legal Events

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

Ref document number: 09784890

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09784890

Country of ref document: EP

Kind code of ref document: A1