US20030147882A1 - Methods for amyloid removal using anti-amyloid antibodies - Google Patents

Methods for amyloid removal using anti-amyloid antibodies Download PDF

Info

Publication number
US20030147882A1
US20030147882A1 US09/316,387 US31638799A US2003147882A1 US 20030147882 A1 US20030147882 A1 US 20030147882A1 US 31638799 A US31638799 A US 31638799A US 2003147882 A1 US2003147882 A1 US 2003147882A1
Authority
US
United States
Prior art keywords
amyloid
fragment
immunoglobulin
antibody
immunoglobulin polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/316,387
Inventor
Alan Solomon
Rudi Hrncic
Jonathan Stuart Wall
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Tennessee Research Foundation
Original Assignee
University of Tennessee Research Foundation
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 University of Tennessee Research Foundation filed Critical University of Tennessee Research Foundation
Priority to US09/316,387 priority Critical patent/US20030147882A1/en
Assigned to UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE reassignment UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HRNCIC, RUDI, WALL, JONATHAN STUART, SOLOMON, ALAN
Assigned to UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE reassignment UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE'S ADDRESS PREVIOUSLY RECORDED ON REEL 010114, FRAME 0739. Assignors: HRNCIC, RUDI, WALL, JONATHAN STUART, SOLOMON, ALAN
Publication of US20030147882A1 publication Critical patent/US20030147882A1/en
Assigned to UNIVERSITY OF TENNESSEE RESEARCH FOUNDATION reassignment UNIVERSITY OF TENNESSEE RESEARCH FOUNDATION CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF TENNESSEE
Priority to US12/693,085 priority patent/US8105594B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention generally relates to methods for treating amyloid-related diseases. Specifically, the present invention provides therapeutic antibody-related methods to effect the removal of amyloid fibrils by a patient's own immunophagocytic system.
  • Amyloidosis refers to the pathological deposition of proteins in the form of congophilic, green birefringent fibrils, when congo red-stained, either dispersed or in the form of localized amyloidomas. Such deposits are symptomatic of several diseases, for example Alzheimer's Disease, inflammation-associated amyloid, type II diabetes, bovine spongiform encephalopathy (BSE), Creutzfeld-Jakob disease (CJD), scrapie and primary amyloidosis.
  • BSE bovine spongiform encephalopathy
  • CJD Creutzfeld-Jakob disease
  • Amyloidoses are generally categorized into three groups: major systemic amyloidoses, major localized amyloidoses, and miscellaneous amyloidoses.
  • Major systemic amyloidoses include: chronic inflammatory conditions (e.g., tuberculosis, osteomyelitis, etc.); non-infectious conditions such as juvenile rheumatoid arthritis, ankylosing spondylitis and Crohn's disease, etc.; familial Mediterranean Fever, plasma cell dyscrasia (primary amyloidosis) and various familial polyneuropathies and cardiomyopathies.
  • Major localized amyloidoses include: chronic dialysis usually for greater than 8 years, Alzheimer's disease, Down syndrome, Hereditary cerebral hemorrhage (Dutch), and non-traumatic cerebral hemorrhage of the elderly. Miscellaneous amyloidoses include: familial polyneuropathy (Iowa), familial amyloidosis (Finnish), hereditary cerebral hemorrhage (Icelandic), CJD, Medullary carcinoma of the thyroid, atrial amyloid, and diabetes mellitus (insulinomas). Other amyloidoses include those referenced in Louis W. Heck, “The Amyloid Diseases” in Cecil's Textbook of Medicine 1504-6 (W. B. Saunders & Co., Philadelphia, Pa.; 1996).
  • Transmissible spongiform encephalopathies which cause CJD and Gerstmann-Strässler-Scheinker (GSS) disease are described by B. Chesebro et al., “Transmissible Spongiform Encephalopathies: A Brief Introduction” in Field's Virology 2845-49 (3rd Edition; Raven Publishers, Philadelphia, Pa.; 1996) and in D. C. Gajdusek, “Infectious amyloids: Subacute Spongiform Encephalopathies as Transmissible Cerebral Amyloidoses,” 2851-2900 in Fields Virology (1996). Many of these diseases are likely mediated by prions, an infectious protein. See S. B.
  • Prusiner “Prions” in Fields Virology 2901-50 (1996) and the references contained therein.
  • amyloidosis very rarely do patients with clinically proven amyloidosis spontaneously achieve complete remission, perhaps because the amyloid fibrils themselves are non-immunogenic.
  • therapies for amyloidosis have been investigated, such as high-dose chemotherapy, steroids, iodinated doxorubicin, and stem cell replacement therapy.
  • Familial-Mediterranean amyloidosis has drug treatment (with colchicine) been shown to be effective.
  • mAbs monoclonal antibodies
  • the present inventors have discovered new methods of treating amyloid-related diseases and conditions. These methods exploit the opsonizing effect of mAbs directed toward the protein constituents of amyloid.
  • the present invention includes a method of treating a patient having an amyloid-associated disease comprising the step of administering to the patient a therapeutically effective dose of at least one immunoglobulin polypeptide, or fragments thereof, together with a pharmaceutically acceptable carrier; wherein the immunoglobulin polypeptide or fragment thereof, may be a substantially purified immunoglobulin polypeptide that binds to a human amyloid fibril, wherein binding of the polypeptide opsonizes the amyloid fibril.
  • the present invention relates to the use of any one of, or a combination of, the three monoclonal antibodies discussed below. These antibodies have general anti-amyloid binding properties and provide an extrinsic opsonizing reagent that activates a patient's own cellular immune clearance mechanism.
  • FIGS. 1A and 1B are reproduced photographs of a Balb/c mouse just after an injection of amyloid is made ( 1 A) and 14 days after the injection ( 1 B). The injection site was shaved to better illustrate the “hump” caused by the injection of the amyloid material.
  • FIGS. 2 A- 2 B are reproduced photographs of human neutrophils (multi-lobed nuclei) adhering to human amyloid opsonized in vitro.
  • FIGS. 3 A- 3 D are reproduced photographs of immunohistochemically stained amyloid-laden tissue samples (20X magnification).
  • FIG. 3A is a tissue sample from a patient with ⁇ 1 amyloidosis stained with Congo red; the amyloid deposits, viewed under polarized light, appear as blue-green particles.
  • FIG. 3B is a tissue sample stained with alkaline phosphatase after labeling with anti- ⁇ I (57-18-H12) mAb.
  • FIG. 3C is a tissue sample stained as in FIG. 3B, but with anti- ⁇ IV (11-1F4) mAb.
  • FIG. 3D is a tissue sample stained as in FIG. 3B, but with anti- ⁇ VIII (31-8c7) mAb.
  • FIG. 4 is a reproduced photograph showing a fluoresceinated (FITC) ⁇ 4 mAb bound to human amyloid implanted into a Balb/c mouse. The mAb was injected into the thigh of the mouse. The amyloidoma was excised 72 hours post injection and viewed using an epifluorescence microscope (20X magnification).
  • FITC fluoresceinated
  • the present invention utilizes immunoglobulin polypeptides to modulate and to enhance the degradation and removal of undesired deposits of amyloid fibrils in a host or patient. It is envisioned that the invention will be used, for example, to treat humans suffering from a disease or condition characterized by an undesired deposition of amyloid fibrils. Without intending to be bound by any particular mechanism of action, it is believed that the administration of immunoglobulin peptides according to the present invention opsonize the deposited amyloid fibrils in a patient suffering from amyloidosis, thereby assisting in their removal from the patient by the patients' own immune system. It is believed that the patient's immune system alone is unable to remove the amyloid fibrils in conditions modulated by amyloid fibrils without such a therapeutic intervention, presumably because the amyloid fibrils are themselves relatively non-immunogenic.
  • a therapeutically effective dose of immunoglobulin polypeptide or fragment thereof according to the present invention is administered together with a pharmaceutically suitable carrier or excipient.
  • a pharmaceutically suitable carrier or excipient Upon the binding or adhering of such immunoglobulin polypeptides to undesired deposits of amyloid fibrils, the latter are believed to be opsonized.
  • compositions of the present invention can be carried out in dosages and by administration protocols known to those skilled in the art for the administration of other therapeutic antibody products. These parameters may be selected and/or optimized by the physician treating a particular patient.
  • a therapeutically effective dose of a pharmaceutical formulation of the present invention should deliver a quantity of anti-amyloid immunoglobulin polypeptide sufficient to substantially inhibit the undesired deposition of amyloid fibrils or to substantially inhibit the rate of any undesired deposition of amyloid fibrils. More preferably, the formulations should reduce the overall burden of deposited amyloid fibrils in a patient. Further, administration of such formulations should begin shortly after diagnosis of amyloidosis and continue until symptoms are substantially abated and for a period thereafter. In well established cases of disease, loading doses followed by maintenance doses may be required.
  • polypeptide indicates that the polypeptide or protein in question is at least about 30% identical to an entire naturally occurring protein or a portion thereof, usually at least about 70% identical, and preferably at least about 95% identical.
  • substantially purified protein will typically comprise over about 85% to 90% of a protein sample, more usually about 95%, and preferably will be over about 99% pure. Protein purity or homogeneity may be indicated by a number of means well known in the art, such a polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band on a polyacrylamide gel upon staining. For certain purposes high resolution will be needed and HPLC or a similar means for purification utilized.
  • Proteins may be purified to substantial homogeneity by standard techniques well known in the art, including selective precipitation with such substances as ammonium sulfate, column chromatography, immunopurification methods, and others. See, for instance, Scopes, Protein Purification: Principles and Practice, Springer-Verlag: New York (1982), which is incorporated herein by reference.
  • Antibody purification techniques are well known in the art. Harlow et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1988), 288-318, which is incorporated herein by reference, describes, for example, purification using ammonium sulfate precipitation, caprlic acid, DEAE, hydroxyapatite chromatography, gel filtration chromatography, protein A beads, and immunoaffinity.
  • Nucleic acids may be DNA or RNA.
  • the term “substantial identity” indicates that the sequences of two nucleic acids, or designated portions thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides.
  • nucleic acid sequence identity exists when a nucleic acid segment will hybridize under selective hybridization conditions, to a complement of another nucleic acid strand.
  • “Substantially complementary” similarly means that one nucleic acid hybridizes selectively to, or is identical to, another nucleic acid. Typically, selective hybridization will occur when there is at least about 55% identity over a stretch of at least 14-25 nucleotides, preferably at least about 65% identity, more preferably at least about 75%, and most preferably at least about 90% identity. See M. Kanehisa Nucleic Acids Res. 12:203 (1984), which is incorporated herein by reference.
  • Stringent hybridization conditions will typically include salt concentrations of less than about 1 M, more usually less than about 500 mM and preferably less than about 200 mM. Temperature conditions will typically be greater than 22° C., typically greater than about 30° C. and preferably in excess of about 37° C. As other factors may dramatically affect the stringency of hybridization, including base composition and size of the complementary strands, presence of organic solvents and extent of base mismatching, the combination of parameters is more important than the absolute measure of any one alone.
  • isolated or “substantially pure,” when referring to nucleic acids, refer to those that have been purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, and others well known in the art. See, F. Ausubel, et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York (1987), incorporated herein by reference.
  • a nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence.
  • operably linked means that the nucleic acid sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame.
  • nucleic acid manipulation such as subcloning nucleic acid sequences encoding polypeptides into expression vectors, labelling probes, DNA hybridization, and so on are described generally, for example in Sambrook et al., (1989) Molecular Cloning: A Laboratory Manual (2nd ed.), Vols. 1-3, Cold Spring Harbor Laboratory, or Ausubel et al., ed. (1987) op. cit., both of which are incorporated herein by reference.
  • “Expression vectors,” “cloning vectors,” or “vectors” are often plasmids or other nucleic acid molecules that are able to replicate in a chosen host cell. Expression vectors may replicate autonomously, or they may replicate by being inserted into a genome of the host cell, by methods well known in the art. Vectors that replicate autonomously will have an origin of replication or autonomous replicating sequence (“ARS”) that is functional in the chosen host cell(s). Often, it is desirable for a vector to be usable in more than one host cell, e.g., in E. coli for cloning and construction, and in a mammalian cell for expression.
  • ARS autonomous replicating sequence
  • Mammalian cell lines are often used as host cells for the expression of polypeptides derived from eukaryotes. Propagation of mammalian cells in culture is per se well known. See, Tissue Culture, Academic Press, Kruse and Patterson, ed. (1973), incorporated herein by reference. Host cell lines may also include such organisms as bacteria (e.g., E. coli or B. subtilis ), yeast, filamentous fungi, plant cells, or insect cells, among others.
  • bacteria e.g., E. coli or B. subtilis
  • yeast e.g., yeast, filamentous fungi, plant cells, or insect cells, among others.
  • Transformation refers to the introduction of vectors containing the nucleic acids of interest directly into host cells by well-known methods. Transformation methods, which vary depending on the type of host cell, include electroporation; transfection employing calcium chloride, rubidium chloride calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; infection (where the vector is an infectious agent); and other methods. See generally, Sambrook et al., (1989) op. cit. Reference to cells into which the nucleic acids described above have been introduced is meant to also include the progeny of such cells.
  • immunoglobulin polypeptide refers to molecules that are derived from native immunoglobulins (e.g., antibodies) that have specific immunoreactive activity against a particular target, e.g., against amyloid fibrils. Antibodies are typically tetramers of immunoglobulin polypeptides. As used herein, the term “antibody” also refers to a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulin genes include those coding for the light chains, which may be of the kappa or lambda types, and those coding for the heavy chains. Heavy chain types are alpha, gamma, delta, epsilon and mu.
  • the carboxy terminal portions of immunoglobulin heavy and light chains are constant regions, while the amino terminal portions are encoded by the myriad immunoglobulin variable region genes.
  • the variable regions of an immunoglobulin are the portions that provide antigen recognition specificity. In particular, the specificity resides in the complementarity determining regions (“CDRs”), also known as hypervariable regions, of the immunoglobulins.
  • CDRs complementarity determining regions
  • the immunoglobulins may exist in a variety of fragment forms including, for example, Fv, Fab, F(ab′′), F(ab′) 2 , SvFv and other fragments, as well as single chains (e.g., Huston, et al., Proc. Nat. Acad. Sci. U.S.A., 85:5879-5883 (1988) and Bird, et al., Science 242:423-426 (1988), which are incorporated herein by reference).
  • Huston, et al., Proc. Nat. Acad. Sci. U.S.A., 85:5879-5883 (1988) and Bird, et al., Science 242:423-426 (1988) which are incorporated herein by reference.
  • Hood, et al. “Immunology,” Benjamin, N.Y., 2nd ed.
  • Immunoglobulin polypeptide also encompasses a truncated immunoglobulin chain, for example, a chain containing less constant region domains than in the native polypeptide.
  • truncated polypeptides can be produced by standard methods such as introducing a stop codon into the gene sequence 5′ of the domain sequences to be deleted. The truncated polypeptides can then be assembled into truncated antibodies.
  • Antibodies as used herein also include bispecific antibodies which can be produced such as by the methods described in the following references: Glennie et al., J. Immunol., 139:2367-2375 (1987); Segal et al., Biologic Therapy of Cancer Therapy of Cancer Updates 2(4):1-12 (1992); and Shalaby et al., J. Exp. Med. 175:217-225 (1992).
  • “Monoclonal antibodies” may be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see Kohler and Milstein, Eur. J. Immunol. 6:511-519 (1976)). Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art.
  • Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host.
  • Monospecific and bispecific immunoglobulins may also be produced by recombinant techniques in prokaryotic or eukaryotic host cells.
  • “Chimeric” antibodies are encoded by immunoglobulin genes that have been genetically engineered so that the light and heavy chain genes are composed of immunoglobulin gene segments belonging to different species.
  • the variable (V) segments of the genes from a mouse monoclonal antibody may be joined to human constant (C) segments.
  • Such a chimeric antibody is likely to be less antigenic to a human than antibodies with mouse constant regions as well as mouse variable regions.
  • chimeric antibody also refers to an antibody that includes an immunoglobulin that has a human-like framework and in which any constant region present has at least about 85%-90%, and preferably about 95% polypeptide sequence identity to a human immunoglobulin constant region, a so-called “humanized” immunoglobulin (see, for example, PCT Publication WO 90/07861, which is incorporated herein by reference).
  • a so-called “humanized” immunoglobulin see, for example, PCT Publication WO 90/07861, which is incorporated herein by reference.
  • all parts of such a “humanized” immunoglobulin, except possibly the complementarity determining regions (CDRs) are substantially identical to corresponding parts of one or more native human immunoglobulin sequences.
  • framework residues may also be replaced with those within or across species especially if certain framework residues are found to affect the structure of the CDRs.
  • a chimeric antibody may also contain truncated variable or constant regions.
  • framework region refers to those portions of immunoglobulin light and heavy chain variable regions that are relatively conserved (i.e., other than the CDRs) among different immunoglobulins in a single species, as defined by Kabat, et al., (1987); Sequences of Proteins of Immunologic Interest, 4th Ed., U.S. Dept. Health and Human Services, which is incorporated herein by reference).
  • a “human-like framework region” is a framework region that in each existing chain comprises at least about 70 or more amino acid residues, typically 75 to 85 or more residues, identical to those in a human immunoglobulin.
  • variable regions or CDRs for producing the chimeric immunoglobulins of the present invention may be similarly derived from monoclonal antibodies capable of binding to the human type amyloid, and will be produced in any convenient mammalian system, including mice, rats, rabbits, human cell lines, or other vertebrates capable of producing antibodies by well-known methods.
  • Variable regions or CDRs may be produced synthetically, by standard recombinant methods, including polymerase chain reaction (“PER”) or through phage-display libraries.
  • Suitable prokaryotic systems such as bacteria, yeast and phage may be employed.
  • Suitable source cells for the DNA sequences and host cells for immunoglobulin expression and secretion can be obtained from a number of sources, such as the American Type Culture Collection (“Catalogue of Cell Lines and Hybridomas,” Fifth edition (1985) Rockville, Md., U.S.A., which is incorporated herein by reference).
  • modified immunoglobulins can be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art.
  • modifications of the genes may be readily accomplished by a variety of well-known techniques, such as PCR and site-directed mutagenesis (see, Gillman and Smith, Gene 8:81-97 (1979) and S. Roberts et al., Nature 328:731-734 (1987), both of which are incorporated herein by reference).
  • polypeptide fragments comprising only a portion of the primary immunoglobulin structure may be produced.
  • Immunoglobulin genes in whole or in part, may also be combined with functional regions from other genes (e.g., enzymes), or with other molecules such as toxins, labels and targeting moieties to produce fusion proteins (e.g., “immunotoxins”) having novel properties. In these cases of gene fusion, the two components are present within the same polypeptide chain.
  • the immunoglobulin or fragment thereof may be chemically bonded to the toxin or label by any of a variety of well-known chemical procedures.
  • the linkage may be by way of heterobifunctional cross-linkers, e.g., SPDP, carbodiimide, glutaraldehyde, or the like.
  • Suitable labels include, for example, radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescers, chemiluminescers, magnetic particles. See, for examples of patents teaching the use of such labels, U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241, all of which are incorporated by reference.
  • Immunotoxins including single chain molecules, may also be produced by recombinant means. Production of various immunotoxins is well-known with the art, and methods can be found, for example in “Monoclonal Antibody-Toxin Conjugates: Aiming the Magic Bullet,” Thorpe et al., Monoclonal Antibodies in Clinical Medicine, Academic Press, pp. 168-190 (1982); E. Vitetta, Science (1987) 238:1098-1104; and G. Winter and C. Milstein, Nature (1991) 349:293-299; all incorporated herein by reference.
  • “Opsonize”, as used herein, refers to the binding of an immunoglobulin polypeptide to a particular target, particularly epitopes found on deposits of amyloid fibrils, such that the antibody and targets together are recognized as “foreign” by the host's cellular immune system.
  • the binding of the immunoglobulin of the present invention enhances the phagocytization of the amyloid fibrils.
  • Amyloidosis is intended to refer to any condition that is characterized by the presence of amyloid material. Such material may be in the form of an amyloidoma or more disperse amyloid deposits or fibrils.
  • the pharmaceutical compositions for therapeutic treatment according to the present invention are intended for parenteral, oral or local administration.
  • the pharmaceutical compositions are administered parenterally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly.
  • the blood brain barrier is impermeable to IgG (see U. Bickel et al., 1994 Bioconjug. Chem. 5: 119-25)
  • delivery of antibodies to overcome the blood-brain barrier (BBB) may be achieved through liposomal or micellar delivery of the antibody to the desired site.
  • the agents of this invention can be delivered directly into the cerebrospinal fluid (see for example L. C. Walker et al., 1994 J. Neuropathol. Exp. Neurol.
  • compositions for parenteral administration which comprise a solution of the anti-amyloid immunoglobulin polypeptide dissolved or suspended in a pharmaceutically acceptable carrier, preferably an aqueous carrier.
  • a pharmaceutically acceptable carrier preferably an aqueous carrier.
  • aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine, hyaluronic acid and the like.
  • These compositions may be sterilized by conventional, well known sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • concentration of anti-amyloid immunoglobulin polypeptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 1%, usually at or at least about 10-15% to as much as 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in an accordance with the particular mode of administration selected.
  • Treatment of humans with amyloidosis according to the present invention could also be applied to animals susceptible to amyloidosis, such as cows or chickens.
  • animals susceptible to amyloidosis such as cows or chickens.
  • references to human patients herein apply also to non-human patients.
  • the immunoglobulin polypeptides are preferably anti-amyloid mAbs directed toward an amyloidoma or components or precursors thereof.
  • the mAbs can be raised against IgLC variable region domains or, preferably, against the IgLC subsets ⁇ 1, ⁇ 4, ⁇ 8, or combinations thereof.
  • the administration to humans of immunoglobulin polypeptides that are substantially non-human may elicit anti-antibody responses. Thus, it may be desirable to prepare anti-IgLC immunoglobulin polypeptides of the present invention which are substantially human.
  • substantially human an antibody or binding fragment thereof comprised of amino acid sequences which are at least about 50% human in origin, at least 70 to 80% more preferred, and about 95-99% or more human most preferred, particularly for repeated administrations over a prolonged period as may be necessary to treat established cases of amyloidosis.
  • human antibody is meant to include antibodies of entirely human origin as well as those which are substantially human, unless the context indicates otherwise.
  • Monoclonal antibodies can also be raised against synthetic amyloid fibrils.
  • Recombinant light chain, variable region peptides are isolated and purified in vitro using standard techniques.
  • Synthetic fibrils are then prepared from the peptides using techniques such as those described by Wall et al., “In vitro Immunoglobulin Light Chain Fibrillogenesis,” METHODS IN ENZYMOLOGY, Vol. 309 (In Press).
  • Antibodies are then raised against the synthetic fibrils using standard immunization techniques, typically in mice or rabbits.
  • Monoclonal cell lines secreting anit-fibril antibodies are produced using standard hybridoma techniques.
  • the anti-amyloid immunoglobulin polypeptides of the invention may be prepared by any of a number of well-known techniques. For instance, they may be prepared by immunizing an animal with purified or partially purified human amyloid.
  • the animals immunized can be any one of a variety of species which are capable of immunologically recognizing epitopes characteristic of the human type amyloid extracellular domain, such as murine, lagomorph, equine, etc.
  • Monoclonal antibodies of the invention may be prepared by immortalizing cells comprising nucleic acid sequences which encode immunoglobulin polypeptides or portions thereof that bind specifically to antigenic determinants characteristic of the extracellular domain of the human type amyloid.
  • the immortalization process can be carried out by hybridoma fusion techniques, by viral transformation of antibody-producing lymphocytes, recombinant DNA techniques, or by techniques that combine cell fusion, viral transformation and/or recombinant DNA methodologies.
  • Immunogens to raise the monoclonal antibodies include synthetic amyloid fibrils as described, for example by, A. Lomakin et al., 1997 Proc. Nat'l Acad. Sci. USA 94: 7942-7, which is incorporated herein by reference.
  • human anti-amyloid monoclonal antibodies may be difficult with conventional immortalization techniques, it may be desirable to first make non-human antibodies and then transfer via recombinant DNA techniques the antigen binding regions of the non-human antibodies, e.g., the Fab, complementarity determining regions (CDRs) or hypervariable regions, to human constant regions (Fc) or framework regions as appropriate to produce substantially human molecules.
  • the antigen binding regions of the non-human antibodies e.g., the Fab, complementarity determining regions (CDRs) or hypervariable regions
  • CDRs complementarity determining regions
  • Fc constant regions
  • the desired human immunoglobulin genes or gene segments can be isolated, for example by PCR from human B cells, the DNA cloned into appropriate vectors for expression in eukaryotic cells and the cloned DNA introduced into animals to produce transgenics.
  • Animals suitable for the production of transgenics expressing human immunoglobulin include mice, rats, rabbits and pigs with rodents of transgenics that express human immunoglobulins should preferably have one or more of their endogenous immunoglobulin loci inactivated or “knocked-out” to facilitate identification and isolation of the human antibodies (See e.g., Lonberg, et al. Nature 368:856-859 (1994)).
  • a typical therapeutic chimeric antibody would be a hybrid protein consisting of the variable (V) or antigen-binding domain from a mouse immunoglobulin specific for a human amyloid antigenic determinant, and the constant (C) or effector domain from a human immunoglobulin, although domains from other mammalian species may be used for both variable and constant domains.
  • the therm “chimeric antibody” also refers to antibodies coded for by immunoglobulin genes in which only the CDRs are transferred from the immunoglobulin that specifically recognizes the antigenic determinants, the remainder of the immunoglobulin gene being derived from a human (or other mammalian, as desired) immunoglobulin gene. As discussed before, this type of chimeric antibody is referred to as a “humanized” (in the case of a human immunoglobulin gene being used) antibody. Also considered are recombinant human antibodies that do not contain sequences of another species.
  • the hypervariable regions of the variable domains of the anti-amyloid immunoglobulin polypeptides comprise a related aspect of the invention.
  • the hypervariable regions, or CDRs in conjunction with the framework regions (those portions of immunoglobulin light and heavy chain variable regions that are relatively conserved among different immunoglobulins in a single species), enable the anti-amyloid immunoglobulin polypeptides to recognize and thus bind to human amyloid.
  • the hypervariable regions can be cloned and sequenced.
  • these regions that confer specific recognition of human amyloid can then be cloned into a vector for expression in a host as part of another immunoglobulin molecule or as a fusion protein, e.g., a carrier molecule which functions to enhance immunogenicity of the cloned idiotype.
  • the anti-amyloid immunoglobulin polypeptides of the invention will generally be used intact, or as immunogenic fragments, such Fv, Fab, F(ab′) 2 fragments.
  • the fragments may be obtained from antibodies by conventional techniques, such as by proteolytic digestion of the antibody using, e.g., pepsin, papain or other proteolytic enzymes, or by recombinant DNA techniques in which a gene or portion thereof encoding the desired fragment is cloned or synthesized, and expressed in a variety of hosts.
  • anti-idiotypic antibodies can be produced by using a specific immunoglobulin as an immunogen in accordance with standard techniques. For example, infection or immunization with an amyloid fibril or fragment thereof, induces a neutralizing immunoglobulin, which has on its Fab variable region combining site an image of the amyloid that is unique to that particular immunoglobulin, i.e., an idiotype. Immunization with such an anti-amyloid immunoglobulin induces an anti-idiotype antibody, which has a conformation at its combining site that mimics the structure of the original amyloid antigen. These anti-idiotype antibodies may therefore be used instead of the amyloid antigen. See, for example, Nisonoff (1991) J. Immunol. 147:2429-2438, which is incorporated herein by reference.
  • Human IgLC amyloid was extracted and purified from infected organs obtained during an autopsy.
  • the first experiments involved transplanting 50-200 mg of this amyloid material into a Balb/c mouse.
  • the amyloid mass, or “amyloidoma,” was prepared in sterile PBS by serial sonication and grinding steps in order to produce a fine suspension of amyloid fibrils complete with the accessory molecules found in vivo. This procedure was performed to allow the amyloid to be injected into the mice through a wide-gauge hypodermic needle.
  • amyloid material equivalent to 10% of the body weight of the animal, was injected into mice (under anesthetic) between the scapula, which resulted in a large mass being visible (see FIG. 1A).
  • the mouse required 15-18 days to achieve the complete removal of the amyloidoma (see FIG. 1B), after which the animal appeared healthy and lived a normal life span.
  • the removal of the amyloidoma was determined subjectively by the experimenter; by simply palpating the injection site, an amyloidoma, like a hard pea, can be easily felt under the skin.
  • FIGS. 2A and 2B show human neutrophils adhering to human amyloid after the amyloid was treated with mouse anti-human IgLC mAbs. This shows that the mouse mAb can bind to human amyloid as well as attract human neutrophils.
  • a Balb/c mouse was injected with 50 mg HIG amyloid and left for 1 week, after which it was bled by tail-vein clipping.
  • the blood was spun down at 1500 rpm and the cells removed by aspiration.
  • the plasma was stored at 4° C. until used.
  • Another preparation of HIG amyloid (100 mg) was prepared by suspending in sterile PBS to which was added 1 ml of plasma from the previous mouse. This preparation was then injected into a second mouse (Balb/c) and the amyloid was removed in 4 days.
  • FIGS. 3 A- 3 D show that anti- ⁇ 1 binds to the ⁇ 1 amyloid and, surprisingly, that the anti- ⁇ 4 reacts with the ⁇ 1 amyloid, suggesting an amyloid epitope that these antibodies may recognize. Additionally, the anti- ⁇ 4 reacts with ⁇ -containing amyloid (not illustrated). This is an example of cross-isotype reactivity.
  • the results from the ELISA and the immunohistochemistry were not always consistent. This is likely due to the inherent difference in what you are looking at, i.e., ELISA is a liquid phase binding assay using extracted purified amyloid, whereas immunohistochemistry is performed on fixed tissue sections on a slide.
  • the ⁇ 1 and ⁇ 4 reagents resulted in the complete removal by the host of most amyloid fibril species tested within 7 days (as little as 4 days for certain sources of amyloid).
  • FIG. 4 shows fluoresceinated ⁇ 4 mAb binding to human amyloid.
  • the ⁇ 8 reagent which is reactive in certain instances in both in vitro studies (above), increased the resolution of amyloidomas by up to about 10% in in vivo experiments.
  • Human amyloid was isolated from a patient with inflammation-associated, AA-amyloid and prepared for injection into Balb/C mice by repeated sonication and grinding in order to permit its injection into the mouse (see Example 1). Immediately after the injection of 100 mg of human AA-amyloid extract, the mice were treated with 100 ⁇ g of ⁇ 4 mAb, anti-AA mAb, no mAb and non-specific control mAb (anti-free ⁇ ). Complete resolution of the material was observed with 48 hours in the animals that had been treated with the ⁇ 4 and anti-AA mAbs. In contrast, the control animals had a large mass of amyloid remaining at the site of injection.
  • Synthetic amyloid fibrils were prepared in vitro and used as an immunogen in mice to produce a first generation of anti-amyloid fibril mAbs. Briefly, recombinant ⁇ 6-light chain, variable region peptides were produced, isolated and purified using a bacterial expression system and standard protein purification techniques. Synthetic fibrils were prepared from these peptides by extended periods of agitation in solution as described, for example, in Wall et al., “In vitro Immunoglobulin Light Chain Fibrillogenesis,” METHODS IN ENZYMOLOGY, Vol. 309 (In Press), which is incorporated herein by reference in its entirety. Fibrils were concentrated by centrifugation at 17,000 ⁇ g for 20 minutes at room temperature.
  • the concentrated fibrils were then used to immunize Balb/c mice over a period of several weeks. Monoclonal cell lines secreting anti-fibril antibodies were produced using standard hybridoma techniques. The resultant antibodies have demonstrable anti-fibril activity based upon ELISA assays, described in Example 3. These antibodies reacted with 99% of all human IgLC amyloid extracts tested to date irrespective of the nature of the isotype or subgroup of the precursor protein when tested by ELISA. Similarly, the antibodies reacted in an ELISA format with isolated murine AA-amyloid and synthetic fibrils composed of a peptide derived from the Alzheimer's protein A ⁇ [A ⁇ (25-35)].

Abstract

Methods and related immunoglobulin peptides and fragments thereof are disclosed that enhance the cell-mediated immune response of a patient to deposits of amyloid fibrils. These methods exploit the opsonizing effect of antibodies directed toward amyloid material or its component parts.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional application No. 60/086,198, filed May 21, 1998, which is herein incorporated by reference in its entirety.[0001]
  • FEDERAL SUPPORT
  • [0002] This invention was made with government support under Grant No. 2 R01 CA 20056, awarded by The National Institutes of Health. Thus, the government may have certain rights in this invention.
  • TECHNICAL FIELD
  • The present invention generally relates to methods for treating amyloid-related diseases. Specifically, the present invention provides therapeutic antibody-related methods to effect the removal of amyloid fibrils by a patient's own immunophagocytic system. [0003]
  • BACKGROUND OF THE INVENTION
  • Amyloidosis refers to the pathological deposition of proteins in the form of congophilic, green birefringent fibrils, when congo red-stained, either dispersed or in the form of localized amyloidomas. Such deposits are symptomatic of several diseases, for example Alzheimer's Disease, inflammation-associated amyloid, type II diabetes, bovine spongiform encephalopathy (BSE), Creutzfeld-Jakob disease (CJD), scrapie and primary amyloidosis. [0004]
  • Amyloidoses are generally categorized into three groups: major systemic amyloidoses, major localized amyloidoses, and miscellaneous amyloidoses. Major systemic amyloidoses include: chronic inflammatory conditions (e.g., tuberculosis, osteomyelitis, etc.); non-infectious conditions such as juvenile rheumatoid arthritis, ankylosing spondylitis and Crohn's disease, etc.; familial Mediterranean Fever, plasma cell dyscrasia (primary amyloidosis) and various familial polyneuropathies and cardiomyopathies. Major localized amyloidoses include: chronic dialysis usually for greater than 8 years, Alzheimer's disease, Down syndrome, Hereditary cerebral hemorrhage (Dutch), and non-traumatic cerebral hemorrhage of the elderly. Miscellaneous amyloidoses include: familial polyneuropathy (Iowa), familial amyloidosis (Finnish), hereditary cerebral hemorrhage (Icelandic), CJD, Medullary carcinoma of the thyroid, atrial amyloid, and diabetes mellitus (insulinomas). Other amyloidoses include those referenced in Louis W. Heck, “The Amyloid Diseases” in Cecil's Textbook of Medicine 1504-6 (W. B. Saunders & Co., Philadelphia, Pa.; 1996). [0005]
  • Transmissible spongiform encephalopathies which cause CJD and Gerstmann-Strässler-Scheinker (GSS) disease are described by B. Chesebro et al., “Transmissible Spongiform Encephalopathies: A Brief Introduction” in Field's Virology 2845-49 (3rd Edition; Raven Publishers, Philadelphia, Pa.; 1996) and in D. C. Gajdusek, “Infectious amyloids: Subacute Spongiform Encephalopathies as Transmissible Cerebral Amyloidoses,” 2851-2900 in Fields Virology (1996). Many of these diseases are likely mediated by prions, an infectious protein. See S. B. Prusiner, “Prions” in Fields Virology 2901-50 (1996) and the references contained therein. The inherited forms of amyloidoses as described on Online Mendelian Inheritance in Man (OMIM) “www.ncbi.nlm.nih.gov/htbin-post/Omim/dispmim?” Each of the above is incorporated herein by reference. [0006]
  • Very rarely do patients with clinically proven amyloidosis spontaneously achieve complete remission, perhaps because the amyloid fibrils themselves are non-immunogenic. Various therapies for amyloidosis have been investigated, such as high-dose chemotherapy, steroids, iodinated doxorubicin, and stem cell replacement therapy. However, in only one type of amyloid disease, Familial-Mediterranean amyloidosis, has drug treatment (with colchicine) been shown to be effective. [0007]
  • The use of monoclonal antibodies (mAbs) to induce or modulate the immunological removal of an otherwise unrecognized entity is known. mAbs have been successfully used in treating non-Hodgkins lymphoma and breast cancer, for example. [0008]
  • Previously, a variety of studies have characterized antibodies that bind to amyloid proteins or amyloid fibrils. See, for example, U.S. Pat. Nos. 5,714,471; 5,693,478; 5,688,651; 5,652,092; 5,593,846; 5,536,640; 5,385,915; 5,348,963; 5,270,165; 5,262,332; 5,262,303; 5,164,295; and 4,782,014. In addition, several publications have suggested that anti-amyloid antibodies might be useful for studying the progression of beta-amyloidosis and for various therapeutic options. See, for example, Bellottii et al., [0009] Scand. J. Immunol. (1992) 36(4):607-615; Bellotti et al., Ren. Fail. (1993) 15(3):365-371; Walker et al. J. Neuropathol. Exp. Neurol. (1994) 53(4):377-383; and Bickel et al., Bioconjug. Chem. (1994) 5(2):119-125. However, no therapeutic antibody has been demonstrated to halt or reverse the deposition of amyloid fibrils in a patient. Thus, a need exists for a method for treating amyloidoses using antibody formulations containing antibodies that bind to amyloid fibrils.
  • SUMMARY OF THE INVENTION
  • The present inventors have discovered new methods of treating amyloid-related diseases and conditions. These methods exploit the opsonizing effect of mAbs directed toward the protein constituents of amyloid. [0010]
  • The present invention includes a method of treating a patient having an amyloid-associated disease comprising the step of administering to the patient a therapeutically effective dose of at least one immunoglobulin polypeptide, or fragments thereof, together with a pharmaceutically acceptable carrier; wherein the immunoglobulin polypeptide or fragment thereof, may be a substantially purified immunoglobulin polypeptide that binds to a human amyloid fibril, wherein binding of the polypeptide opsonizes the amyloid fibril. [0011]
  • In particular, the present invention relates to the use of any one of, or a combination of, the three monoclonal antibodies discussed below. These antibodies have general anti-amyloid binding properties and provide an extrinsic opsonizing reagent that activates a patient's own cellular immune clearance mechanism.[0012]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A and 1B. FIGS. 1A and 1B are reproduced photographs of a Balb/c mouse just after an injection of amyloid is made ([0013] 1A) and 14 days after the injection (1B). The injection site was shaved to better illustrate the “hump” caused by the injection of the amyloid material.
  • FIGS. [0014] 2A-2B. FIGS. 2A and 2B are reproduced photographs of human neutrophils (multi-lobed nuclei) adhering to human amyloid opsonized in vitro.
  • FIGS. [0015] 3A-3D. FIGS. 3A-3D are reproduced photographs of immunohistochemically stained amyloid-laden tissue samples (20X magnification). FIG. 3A is a tissue sample from a patient with κ1 amyloidosis stained with Congo red; the amyloid deposits, viewed under polarized light, appear as blue-green particles. FIG. 3B is a tissue sample stained with alkaline phosphatase after labeling with anti-κI (57-18-H12) mAb. FIG. 3C is a tissue sample stained as in FIG. 3B, but with anti-κIV (11-1F4) mAb. FIG. 3D is a tissue sample stained as in FIG. 3B, but with anti-λVIII (31-8c7) mAb.
  • FIG. 4. FIG. 4 is a reproduced photograph showing a fluoresceinated (FITC) κ4 mAb bound to human amyloid implanted into a Balb/c mouse. The mAb was injected into the thigh of the mouse. The amyloidoma was excised 72 hours post injection and viewed using an epifluorescence microscope (20X magnification).[0016]
  • MODES OF CARRYING OUT THE INVENTION
  • General Description [0017]
  • The present invention utilizes immunoglobulin polypeptides to modulate and to enhance the degradation and removal of undesired deposits of amyloid fibrils in a host or patient. It is envisioned that the invention will be used, for example, to treat humans suffering from a disease or condition characterized by an undesired deposition of amyloid fibrils. Without intending to be bound by any particular mechanism of action, it is believed that the administration of immunoglobulin peptides according to the present invention opsonize the deposited amyloid fibrils in a patient suffering from amyloidosis, thereby assisting in their removal from the patient by the patients' own immune system. It is believed that the patient's immune system alone is unable to remove the amyloid fibrils in conditions modulated by amyloid fibrils without such a therapeutic intervention, presumably because the amyloid fibrils are themselves relatively non-immunogenic. [0018]
  • To treat a patient with amyloidosis, a therapeutically effective dose of immunoglobulin polypeptide or fragment thereof according to the present invention is administered together with a pharmaceutically suitable carrier or excipient. Upon the binding or adhering of such immunoglobulin polypeptides to undesired deposits of amyloid fibrils, the latter are believed to be opsonized. [0019]
  • Single or multiple administrations of the compositions of the present invention can be carried out in dosages and by administration protocols known to those skilled in the art for the administration of other therapeutic antibody products. These parameters may be selected and/or optimized by the physician treating a particular patient. [0020]
  • Preferably, a therapeutically effective dose of a pharmaceutical formulation of the present invention should deliver a quantity of anti-amyloid immunoglobulin polypeptide sufficient to substantially inhibit the undesired deposition of amyloid fibrils or to substantially inhibit the rate of any undesired deposition of amyloid fibrils. More preferably, the formulations should reduce the overall burden of deposited amyloid fibrils in a patient. Further, administration of such formulations should begin shortly after diagnosis of amyloidosis and continue until symptoms are substantially abated and for a period thereafter. In well established cases of disease, loading doses followed by maintenance doses may be required. [0021]
  • Definitions [0022]
  • The terms “peptide,” “polypeptide” or “protein” are used interchangeably herein. The term “substantial identity,” when referring to polypeptides, indicates that the polypeptide or protein in question is at least about 30% identical to an entire naturally occurring protein or a portion thereof, usually at least about 70% identical, and preferably at least about 95% identical. [0023]
  • As used herein, the terms “isolated,” “substantially pure” and “substantially homogenous” are used interchangeably and describe a protein that has been separated from components which naturally accompany it. A substantially purified protein will typically comprise over about 85% to 90% of a protein sample, more usually about 95%, and preferably will be over about 99% pure. Protein purity or homogeneity may be indicated by a number of means well known in the art, such a polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band on a polyacrylamide gel upon staining. For certain purposes high resolution will be needed and HPLC or a similar means for purification utilized. [0024]
  • Proteins may be purified to substantial homogeneity by standard techniques well known in the art, including selective precipitation with such substances as ammonium sulfate, column chromatography, immunopurification methods, and others. See, for instance, Scopes, [0025] Protein Purification: Principles and Practice, Springer-Verlag: New York (1982), which is incorporated herein by reference.
  • Antibody purification techniques are well known in the art. Harlow et al., [0026] Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1988), 288-318, which is incorporated herein by reference, describes, for example, purification using ammonium sulfate precipitation, caprlic acid, DEAE, hydroxyapatite chromatography, gel filtration chromatography, protein A beads, and immunoaffinity.
  • Nucleic acids, as used herein, may be DNA or RNA. When referring to nucleic acids, the term “substantial identity” indicates that the sequences of two nucleic acids, or designated portions thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides. [0027]
  • Alternatively, substantial nucleic acid sequence identity exists when a nucleic acid segment will hybridize under selective hybridization conditions, to a complement of another nucleic acid strand. [0028]
  • “Substantially complementary” similarly means that one nucleic acid hybridizes selectively to, or is identical to, another nucleic acid. Typically, selective hybridization will occur when there is at least about 55% identity over a stretch of at least 14-25 nucleotides, preferably at least about 65% identity, more preferably at least about 75%, and most preferably at least about 90% identity. See M. Kanehisa [0029] Nucleic Acids Res. 12:203 (1984), which is incorporated herein by reference.
  • Stringent hybridization conditions will typically include salt concentrations of less than about 1 M, more usually less than about 500 mM and preferably less than about 200 mM. Temperature conditions will typically be greater than 22° C., typically greater than about 30° C. and preferably in excess of about 37° C. As other factors may dramatically affect the stringency of hybridization, including base composition and size of the complementary strands, presence of organic solvents and extent of base mismatching, the combination of parameters is more important than the absolute measure of any one alone. [0030]
  • “Isolated” or “substantially pure,” when referring to nucleic acids, refer to those that have been purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, and others well known in the art. See, F. Ausubel, et al., ed. [0031] Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York (1987), incorporated herein by reference.
  • A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence. Generally, operably linked means that the nucleic acid sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame. [0032]
  • Techniques for nucleic acid manipulation, such as subcloning nucleic acid sequences encoding polypeptides into expression vectors, labelling probes, DNA hybridization, and so on are described generally, for example in Sambrook et al., (1989) [0033] Molecular Cloning: A Laboratory Manual (2nd ed.), Vols. 1-3, Cold Spring Harbor Laboratory, or Ausubel et al., ed. (1987) op. cit., both of which are incorporated herein by reference.
  • “Expression vectors,” “cloning vectors,” or “vectors” are often plasmids or other nucleic acid molecules that are able to replicate in a chosen host cell. Expression vectors may replicate autonomously, or they may replicate by being inserted into a genome of the host cell, by methods well known in the art. Vectors that replicate autonomously will have an origin of replication or autonomous replicating sequence (“ARS”) that is functional in the chosen host cell(s). Often, it is desirable for a vector to be usable in more than one host cell, e.g., in [0034] E. coli for cloning and construction, and in a mammalian cell for expression.
  • Mammalian cell lines are often used as host cells for the expression of polypeptides derived from eukaryotes. Propagation of mammalian cells in culture is per se well known. See, [0035] Tissue Culture, Academic Press, Kruse and Patterson, ed. (1973), incorporated herein by reference. Host cell lines may also include such organisms as bacteria (e.g., E. coli or B. subtilis), yeast, filamentous fungi, plant cells, or insect cells, among others.
  • “Transformation” refers to the introduction of vectors containing the nucleic acids of interest directly into host cells by well-known methods. Transformation methods, which vary depending on the type of host cell, include electroporation; transfection employing calcium chloride, rubidium chloride calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; infection (where the vector is an infectious agent); and other methods. See generally, Sambrook et al., (1989) op. cit. Reference to cells into which the nucleic acids described above have been introduced is meant to also include the progeny of such cells. [0036]
  • As used herein, “immunoglobulin polypeptide” refers to molecules that are derived from native immunoglobulins (e.g., antibodies) that have specific immunoreactive activity against a particular target, e.g., against amyloid fibrils. Antibodies are typically tetramers of immunoglobulin polypeptides. As used herein, the term “antibody” also refers to a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulin genes include those coding for the light chains, which may be of the kappa or lambda types, and those coding for the heavy chains. Heavy chain types are alpha, gamma, delta, epsilon and mu. The carboxy terminal portions of immunoglobulin heavy and light chains are constant regions, while the amino terminal portions are encoded by the myriad immunoglobulin variable region genes. The variable regions of an immunoglobulin are the portions that provide antigen recognition specificity. In particular, the specificity resides in the complementarity determining regions (“CDRs”), also known as hypervariable regions, of the immunoglobulins. [0037]
  • The immunoglobulins may exist in a variety of fragment forms including, for example, Fv, Fab, F(ab″), F(ab′)[0038] 2, SvFv and other fragments, as well as single chains (e.g., Huston, et al., Proc. Nat. Acad. Sci. U.S.A., 85:5879-5883 (1988) and Bird, et al., Science 242:423-426 (1988), which are incorporated herein by reference). (See, generally, Hood, et al., “Immunology,” Benjamin, N.Y., 2nd ed. (1984), and Hunkapiller and Hood, Nature, 323:15-16 (1986), which are incorporated herein by reference). Single-chain antibodies, in which genes for a heavy chain and a light chain are combined into a single coding sequence, may also be used. Immunoglobulin polypeptide also encompasses a truncated immunoglobulin chain, for example, a chain containing less constant region domains than in the native polypeptide. Such truncated polypeptides can be produced by standard methods such as introducing a stop codon into the gene sequence 5′ of the domain sequences to be deleted. The truncated polypeptides can then be assembled into truncated antibodies. Antibodies as used herein also include bispecific antibodies which can be produced such as by the methods described in the following references: Glennie et al., J. Immunol., 139:2367-2375 (1987); Segal et al., Biologic Therapy of Cancer Therapy of Cancer Updates 2(4):1-12 (1992); and Shalaby et al., J. Exp. Med. 175:217-225 (1992).
  • “Monoclonal antibodies” may be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see Kohler and Milstein, [0039] Eur. J. Immunol. 6:511-519 (1976)). Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host.
  • Monospecific and bispecific immunoglobulins may also be produced by recombinant techniques in prokaryotic or eukaryotic host cells. [0040]
  • “Chimeric” antibodies are encoded by immunoglobulin genes that have been genetically engineered so that the light and heavy chain genes are composed of immunoglobulin gene segments belonging to different species. For example, the variable (V) segments of the genes from a mouse monoclonal antibody may be joined to human constant (C) segments. Such a chimeric antibody is likely to be less antigenic to a human than antibodies with mouse constant regions as well as mouse variable regions. [0041]
  • As used herein, the term chimeric antibody also refers to an antibody that includes an immunoglobulin that has a human-like framework and in which any constant region present has at least about 85%-90%, and preferably about 95% polypeptide sequence identity to a human immunoglobulin constant region, a so-called “humanized” immunoglobulin (see, for example, PCT Publication WO 90/07861, which is incorporated herein by reference). Hence, all parts of such a “humanized” immunoglobulin, except possibly the complementarity determining regions (CDRs), are substantially identical to corresponding parts of one or more native human immunoglobulin sequences. Where necessary, framework residues may also be replaced with those within or across species especially if certain framework residues are found to affect the structure of the CDRs. A chimeric antibody may also contain truncated variable or constant regions. [0042]
  • The term “framework region,” as used herein, refers to those portions of immunoglobulin light and heavy chain variable regions that are relatively conserved (i.e., other than the CDRs) among different immunoglobulins in a single species, as defined by Kabat, et al., (1987); [0043] Sequences of Proteins of Immunologic Interest, 4th Ed., U.S. Dept. Health and Human Services, which is incorporated herein by reference). As used herein, a “human-like framework region” is a framework region that in each existing chain comprises at least about 70 or more amino acid residues, typically 75 to 85 or more residues, identical to those in a human immunoglobulin.
  • Human constant region DNA sequences can be isolated in accordance with well-known procedures from a variety of human cells, but preferably from immortalized B-cells. The variable regions or CDRs for producing the chimeric immunoglobulins of the present invention may be similarly derived from monoclonal antibodies capable of binding to the human type amyloid, and will be produced in any convenient mammalian system, including mice, rats, rabbits, human cell lines, or other vertebrates capable of producing antibodies by well-known methods. Variable regions or CDRs may be produced synthetically, by standard recombinant methods, including polymerase chain reaction (“PER”) or through phage-display libraries. For phage display methods, see for example, McCafferty et al., [0044] Nature 348:552-554 (1990); Clackson et al., Nature 352:624-628 and Marks et al., Biotechnology 11:1145-1149 (1993). Suitable prokaryotic systems such as bacteria, yeast and phage may be employed.
  • Suitable source cells for the DNA sequences and host cells for immunoglobulin expression and secretion can be obtained from a number of sources, such as the American Type Culture Collection (“Catalogue of Cell Lines and Hybridomas,” Fifth edition (1985) Rockville, Md., U.S.A., which is incorporated herein by reference). [0045]
  • In addition to the chimeric and “humanized” immunoglobulins specifically described herein, other substantially identical modified immunoglobulins can be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art. In general, modifications of the genes may be readily accomplished by a variety of well-known techniques, such as PCR and site-directed mutagenesis (see, Gillman and Smith, [0046] Gene 8:81-97 (1979) and S. Roberts et al., Nature 328:731-734 (1987), both of which are incorporated herein by reference).
  • Alternatively, polypeptide fragments comprising only a portion of the primary immunoglobulin structure may be produced. For example, it may be desirable to produce immunoglobulin polypeptide fragments that possess one or more immunoglobulin activities in addition to, or other than, antigen recognition (e.g., complement fixation). [0047]
  • Immunoglobulin genes, in whole or in part, may also be combined with functional regions from other genes (e.g., enzymes), or with other molecules such as toxins, labels and targeting moieties to produce fusion proteins (e.g., “immunotoxins”) having novel properties. In these cases of gene fusion, the two components are present within the same polypeptide chain. Alternatively, the immunoglobulin or fragment thereof may be chemically bonded to the toxin or label by any of a variety of well-known chemical procedures. For example, when the label or cytotoxic agent is a protein and the second component is an intact immunoglobulin, the linkage may be by way of heterobifunctional cross-linkers, e.g., SPDP, carbodiimide, glutaraldehyde, or the like. [0048]
  • Suitable labels include, for example, radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescers, chemiluminescers, magnetic particles. See, for examples of patents teaching the use of such labels, U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241, all of which are incorporated by reference. [0049]
  • Immunotoxins, including single chain molecules, may also be produced by recombinant means. Production of various immunotoxins is well-known with the art, and methods can be found, for example in “Monoclonal Antibody-Toxin Conjugates: Aiming the Magic Bullet,” Thorpe et al., [0050] Monoclonal Antibodies in Clinical Medicine, Academic Press, pp. 168-190 (1982); E. Vitetta, Science (1987) 238:1098-1104; and G. Winter and C. Milstein, Nature (1991) 349:293-299; all incorporated herein by reference.
  • Additional techniques for preparing immunoglobulins and immunoglobulin fragments are described in V. S. Malik et al, [0051] Antibody Techniques (Academic Press, 1994); C. A. K. Borrebaeck, Antibody Engineering: Breakthroughs in Molecular Biology (Oxford Univ. Press, 1995); and P. J. Delves et al., Antibody Production: Essential Techniques (John Wiley & Sons, 1997), which are incorporated herein by reference.
  • “Opsonize”, as used herein, refers to the binding of an immunoglobulin polypeptide to a particular target, particularly epitopes found on deposits of amyloid fibrils, such that the antibody and targets together are recognized as “foreign” by the host's cellular immune system. In other words the binding of the immunoglobulin of the present invention enhances the phagocytization of the amyloid fibrils. [0052]
  • “Amyloidosis”, as used herein, is intended to refer to any condition that is characterized by the presence of amyloid material. Such material may be in the form of an amyloidoma or more disperse amyloid deposits or fibrils. [0053]
  • Pharmaceutical Compositions [0054]
  • The pharmaceutical compositions for therapeutic treatment according to the present invention are intended for parenteral, oral or local administration. Preferably, the pharmaceutical compositions are administered parenterally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly. As the blood brain barrier is impermeable to IgG (see U. Bickel et al., 1994 Bioconjug. Chem. 5: 119-25), delivery of antibodies to overcome the blood-brain barrier (BBB) may be achieved through liposomal or micellar delivery of the antibody to the desired site. Alternatively, the agents of this invention can be delivered directly into the cerebrospinal fluid (see for example L. C. Walker et al., 1994 J. Neuropathol. Exp. Neurol. 53: 377-83). For other delivery mechanisms, refer to P. M. Friden, 1996 U.S. Pat. No. 5,527,527 and W. M. Pardridge, 1991 U.S. Pat. No. 5,004,697. All of the above documents are incorporated herein by reference. [0055]
  • Thus, the invention provides compositions for parenteral administration which comprise a solution of the anti-amyloid immunoglobulin polypeptide dissolved or suspended in a pharmaceutically acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc. [0056]
  • The concentration of anti-amyloid immunoglobulin polypeptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 1%, usually at or at least about 10-15% to as much as 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in an accordance with the particular mode of administration selected. [0057]
  • Without undue experimentation, one of ordinary skill in the art could determine the quantity of immunoglobulin polypeptides that would be effective in adequately opsonizing an amyloidoma. Amounts effective for this use will depend on, e.g., the nature of the anti-amyloid immunoglobulin polypeptide composition, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician. A typical single dose of 0.5 mg/kg could generally be used. It must be kept in an mind that the anti-amyloid immunoglobulin polypeptide and peptide compositions derived therefrom may be employed in serious disease states, that is, life-threatening or potentially life-threatening situations. In such cases it is possible and may be felt desirable by the treating physician to administer substantial excesses of these compositions. Thus, human anti-amyloid monoclonal antibodies or substantially human anti-amyloid receptor monoclonal antibodies of the invention are most preferred under these circumstances. [0058]
  • Treatment of humans with amyloidosis according to the present invention could also be applied to animals susceptible to amyloidosis, such as cows or chickens. Thus, references to human patients herein apply also to non-human patients. [0059]
  • The immunoglobulin polypeptides, as defined herein, are preferably anti-amyloid mAbs directed toward an amyloidoma or components or precursors thereof. The mAbs can be raised against IgLC variable region domains or, preferably, against the IgLC subsets κ1, κ4, λ8, or combinations thereof. The administration to humans of immunoglobulin polypeptides that are substantially non-human may elicit anti-antibody responses. Thus, it may be desirable to prepare anti-IgLC immunoglobulin polypeptides of the present invention which are substantially human. By “substantially human” is meant an antibody or binding fragment thereof comprised of amino acid sequences which are at least about 50% human in origin, at least 70 to 80% more preferred, and about 95-99% or more human most preferred, particularly for repeated administrations over a prolonged period as may be necessary to treat established cases of amyloidosis. As used herein, human antibody is meant to include antibodies of entirely human origin as well as those which are substantially human, unless the context indicates otherwise. [0060]
  • Monoclonal antibodies can also be raised against synthetic amyloid fibrils. Recombinant light chain, variable region peptides are isolated and purified in vitro using standard techniques. Synthetic fibrils are then prepared from the peptides using techniques such as those described by Wall et al., “In vitro Immunoglobulin Light Chain Fibrillogenesis,” METHODS IN ENZYMOLOGY, Vol. 309 (In Press). Antibodies are then raised against the synthetic fibrils using standard immunization techniques, typically in mice or rabbits. Monoclonal cell lines secreting anit-fibril antibodies are produced using standard hybridoma techniques. [0061]
  • The anti-amyloid immunoglobulin polypeptides of the invention may be prepared by any of a number of well-known techniques. For instance, they may be prepared by immunizing an animal with purified or partially purified human amyloid. The animals immunized can be any one of a variety of species which are capable of immunologically recognizing epitopes characteristic of the human type amyloid extracellular domain, such as murine, lagomorph, equine, etc. [0062]
  • Monoclonal antibodies of the invention may be prepared by immortalizing cells comprising nucleic acid sequences which encode immunoglobulin polypeptides or portions thereof that bind specifically to antigenic determinants characteristic of the extracellular domain of the human type amyloid. The immortalization process can be carried out by hybridoma fusion techniques, by viral transformation of antibody-producing lymphocytes, recombinant DNA techniques, or by techniques that combine cell fusion, viral transformation and/or recombinant DNA methodologies. Immunogens to raise the monoclonal antibodies include synthetic amyloid fibrils as described, for example by, A. Lomakin et al., 1997 Proc. Nat'l Acad. Sci. USA 94: 7942-7, which is incorporated herein by reference. [0063]
  • As the generation of human anti-amyloid monoclonal antibodies may be difficult with conventional immortalization techniques, it may be desirable to first make non-human antibodies and then transfer via recombinant DNA techniques the antigen binding regions of the non-human antibodies, e.g., the Fab, complementarity determining regions (CDRs) or hypervariable regions, to human constant regions (Fc) or framework regions as appropriate to produce substantially human molecules. Such methods are generally known in the art and are described in, for example, U.S. Pat. No. 4,816,397, PCT publication WO 90/07861, and EP publications 173494 and 239400, wherein each is incorporated herein by reference. However, completely human antibodies can be produced in transgenic animals. The desired human immunoglobulin genes or gene segments can be isolated, for example by PCR from human B cells, the DNA cloned into appropriate vectors for expression in eukaryotic cells and the cloned DNA introduced into animals to produce transgenics. Animals suitable for the production of transgenics expressing human immunoglobulin include mice, rats, rabbits and pigs with rodents of transgenics that express human immunoglobulins should preferably have one or more of their endogenous immunoglobulin loci inactivated or “knocked-out” to facilitate identification and isolation of the human antibodies (See e.g., Lonberg, et al. [0064] Nature 368:856-859 (1994)).
  • The resulting chimeric antibodies or chimeric immunoglobulin polypeptides that bind to human amyloid are also within the scope of the present invention. A typical therapeutic chimeric antibody would be a hybrid protein consisting of the variable (V) or antigen-binding domain from a mouse immunoglobulin specific for a human amyloid antigenic determinant, and the constant (C) or effector domain from a human immunoglobulin, although domains from other mammalian species may be used for both variable and constant domains. As used herein, the therm “chimeric antibody” also refers to antibodies coded for by immunoglobulin genes in which only the CDRs are transferred from the immunoglobulin that specifically recognizes the antigenic determinants, the remainder of the immunoglobulin gene being derived from a human (or other mammalian, as desired) immunoglobulin gene. As discussed before, this type of chimeric antibody is referred to as a “humanized” (in the case of a human immunoglobulin gene being used) antibody. Also considered are recombinant human antibodies that do not contain sequences of another species. [0065]
  • The hypervariable regions of the variable domains of the anti-amyloid immunoglobulin polypeptides comprise a related aspect of the invention. The hypervariable regions, or CDRs, in conjunction with the framework regions (those portions of immunoglobulin light and heavy chain variable regions that are relatively conserved among different immunoglobulins in a single species), enable the anti-amyloid immunoglobulin polypeptides to recognize and thus bind to human amyloid. The hypervariable regions can be cloned and sequenced. Once identified, these regions that confer specific recognition of human amyloid can then be cloned into a vector for expression in a host as part of another immunoglobulin molecule or as a fusion protein, e.g., a carrier molecule which functions to enhance immunogenicity of the cloned idiotype. [0066]
  • The anti-amyloid immunoglobulin polypeptides of the invention will generally be used intact, or as immunogenic fragments, such Fv, Fab, F(ab′)[0067] 2 fragments. The fragments may be obtained from antibodies by conventional techniques, such as by proteolytic digestion of the antibody using, e.g., pepsin, papain or other proteolytic enzymes, or by recombinant DNA techniques in which a gene or portion thereof encoding the desired fragment is cloned or synthesized, and expressed in a variety of hosts.
  • Those skilled in the art will realize that “anti-idiotypic” antibodies can be produced by using a specific immunoglobulin as an immunogen in accordance with standard techniques. For example, infection or immunization with an amyloid fibril or fragment thereof, induces a neutralizing immunoglobulin, which has on its Fab variable region combining site an image of the amyloid that is unique to that particular immunoglobulin, i.e., an idiotype. Immunization with such an anti-amyloid immunoglobulin induces an anti-idiotype antibody, which has a conformation at its combining site that mimics the structure of the original amyloid antigen. These anti-idiotype antibodies may therefore be used instead of the amyloid antigen. See, for example, Nisonoff (1991) [0068] J. Immunol. 147:2429-2438, which is incorporated herein by reference.
  • The following working examples specifically point out preferred embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure. Other generic configurations will be apparent to one skilled in the art. [0069]
  • EXAMPLE 1 Unassisted Resolution of Human IgLC Amyloid in Murine Host
  • Human IgLC amyloid was extracted and purified from infected organs obtained during an autopsy. The first experiments involved transplanting 50-200 mg of this amyloid material into a Balb/c mouse. The amyloid mass, or “amyloidoma,” was prepared in sterile PBS by serial sonication and grinding steps in order to produce a fine suspension of amyloid fibrils complete with the accessory molecules found in vivo. This procedure was performed to allow the amyloid to be injected into the mice through a wide-gauge hypodermic needle. [0070]
  • The amyloid material, equivalent to 10% of the body weight of the animal, was injected into mice (under anesthetic) between the scapula, which resulted in a large mass being visible (see FIG. 1A). The mouse required 15-18 days to achieve the complete removal of the amyloidoma (see FIG. 1B), after which the animal appeared healthy and lived a normal life span. The removal of the amyloidoma was determined subjectively by the experimenter; by simply palpating the injection site, an amyloidoma, like a hard pea, can be easily felt under the skin. [0071]
  • EXAMPLE 2 Involvement of Both Antibody-Meditated and Cellular Immunity in the Removal of Amyloidomas
  • The involvement of anti-amyloid antibodies in the removal of amyloidomas was shown by screening serum from a mouse previously injected with amyloid material against a sample of the injected material. This was done by Western blot analysis using suitable dilutions of the mouse serum as the primary antibody. It was shown that there were antibodies to every component of the amyloid matrix, i.e., every band on the gel was stained by the mouse serum, even at a 10,000-fold serum dilution (data not shown). [0072]
  • The involvement of a cellular component was demonstrated by in vitro neutrophil binding assays (see FIGS. 2A and 2B) and by using knockout-mutant mouse strains (data not shown). FIGS. 2A and 2B show human neutrophils adhering to human amyloid after the amyloid was treated with mouse anti-human IgLC mAbs. This shows that the mouse mAb can bind to human amyloid as well as attract human neutrophils. [0073]
  • Studies of knockout-mutant mouse strains further support a finding of antibody involvement in amyloid removal. First, scid/scid mice, which lack B and T lymphocytes, were unable to remove an injected amyloidoma even after three months (data not shown). Second, CD18 knockout animals were unable to remove the amyloidoma as rapidly as normal animals. CD18 knockout animals are 97% deficient in CD18, a cell surface integrin found on granulocyte/macrophage lineages. Although these cells cannot leave the circulation, the animals are B and T cell competent and can therefore mount an antibody response. Third, nude mice, which have no white blood cells, were unable to remove the amyloidoma. [0074]
  • Furthermore, amyloid that had been incubated with amyloid-reactive serum from another mouse, when implanted into the second mouse, was removed within 4 days. In this experiment a Balb/c mouse was injected with 50 mg HIG amyloid and left for 1 week, after which it was bled by tail-vein clipping. The blood was spun down at 1500 rpm and the cells removed by aspiration. The plasma was stored at 4° C. until used. Another preparation of HIG amyloid (100 mg) was prepared by suspending in sterile PBS to which was added 1 ml of plasma from the previous mouse. This preparation was then injected into a second mouse (Balb/c) and the amyloid was removed in 4 days. Thus, it was concluded that the process could be sped up by opsonizing the material prior to injection. [0075]
  • EXAMPLE 3 ELISA Screening of IgLC Subsets
  • A systematic study was performed using ELISA techniques to screen a large number of human extracted amyloid samples using mAbs raised against the IgLC subsets (λ1, λ2, λ3, λ4, λ5, λ6, κ1, κ2, κ3, κ4, free κ and λ and total κ and λ). Interestingly, it was found that more often than not, the amyloids tested positive with mAbs specific for their own subtype, the total κ or λ antibodies and a κ1(57-18H12), κ4(11-1F4) and λ8(31-8C7) mAb. These latter three reagents were found to react in a non-subgroup specific manner, i.e., κ1 reacted with amyloids comprised of IgLCs other than κ1; and the other two mAbs exhibit the same quality. This shows that the epitope recognized by these antibodies may be a general feature of amyloid fibrils, indicating the possibility of a shared amyloid epitope that can be targeted. [0076]
  • EXAMPLE 4 Immunochemical Staining
  • Tissue samples from amyloid patients were stained using standard immunochemical techniques and a similar binding phenomenon was observed. FIGS. [0077] 3A-3D show that anti-κ1 binds to the κ1 amyloid and, surprisingly, that the anti-κ4 reacts with the κ1 amyloid, suggesting an amyloid epitope that these antibodies may recognize. Additionally, the anti-κ4 reacts with λ-containing amyloid (not illustrated). This is an example of cross-isotype reactivity. However, the results from the ELISA and the immunohistochemistry were not always consistent. This is likely due to the inherent difference in what you are looking at, i.e., ELISA is a liquid phase binding assay using extracted purified amyloid, whereas immunohistochemistry is performed on fixed tissue sections on a slide.
  • Samples of hybridoma cells that secrete anti-κ1 (57-18-H12 (ATCC Acc. No. ______)), anti-κ4 (11-1F4 (ATCC Acc. No. ______)) and anti-κ8 (31-8c7 (ATCC Acc. No. ______)) monoclonal antibodies were deposited with the American Type Culture Collection (ATCC) on May 21, 1999 in compliance with the Budapest Treaty. [0078]
  • EXAMPLE 5 In Vivo Studies of Anti-IgLC Subgroups
  • 0.1 mg of one of three antibodies—κ1, κ4, or λ8, identified above—was injected into the thigh of a mouse into which amyloid had been introduced in the form of an amyloidoma as described above. The κ1 and κ4 reagents resulted in the complete removal by the host of most amyloid fibril species tested within 7 days (as little as 4 days for certain sources of amyloid). FIG. 4 shows fluoresceinated κ4 mAb binding to human amyloid. [0079]
  • The λ8 reagent, which is reactive in certain instances in both in vitro studies (above), increased the resolution of amyloidomas by up to about 10% in in vivo experiments. [0080]
  • EXAMPLE 6 In Vivo Studies of Anti-IgLC Subgroups
  • Human amyloid was isolated from a patient with inflammation-associated, AA-amyloid and prepared for injection into Balb/C mice by repeated sonication and grinding in order to permit its injection into the mouse (see Example 1). Immediately after the injection of 100 mg of human AA-amyloid extract, the mice were treated with 100 μg of κ4 mAb, anti-AA mAb, no mAb and non-specific control mAb (anti-free κ). Complete resolution of the material was observed with 48 hours in the animals that had been treated with the κ4 and anti-AA mAbs. In contrast, the control animals had a large mass of amyloid remaining at the site of injection. [0081]
  • EXAMPLE 7 Production of Specific Anti-Amyloid Fibril mAbs
  • Synthetic amyloid fibrils were prepared in vitro and used as an immunogen in mice to produce a first generation of anti-amyloid fibril mAbs. Briefly, recombinant λ6-light chain, variable region peptides were produced, isolated and purified using a bacterial expression system and standard protein purification techniques. Synthetic fibrils were prepared from these peptides by extended periods of agitation in solution as described, for example, in Wall et al., “In vitro Immunoglobulin Light Chain Fibrillogenesis,” METHODS IN ENZYMOLOGY, Vol. 309 (In Press), which is incorporated herein by reference in its entirety. Fibrils were concentrated by centrifugation at 17,000×g for 20 minutes at room temperature. [0082]
  • The concentrated fibrils were then used to immunize Balb/c mice over a period of several weeks. Monoclonal cell lines secreting anti-fibril antibodies were produced using standard hybridoma techniques. The resultant antibodies have demonstrable anti-fibril activity based upon ELISA assays, described in Example 3. These antibodies reacted with 99% of all human IgLC amyloid extracts tested to date irrespective of the nature of the isotype or subgroup of the precursor protein when tested by ELISA. Similarly, the antibodies reacted in an ELISA format with isolated murine AA-amyloid and synthetic fibrils composed of a peptide derived from the Alzheimer's protein Aβ [Aβ(25-35)]. [0083]
  • It should be understood that the foregoing discussion and examples merely present a detailed description of certain preferred embodiments. It therefore should be apparent to those of ordinary skill in the art that various modifications and equivalents can be made without departing from the spirit and scope of the invention. All references, articles and patents identified above are herein incorporated by reference in their entirety. [0084]

Claims (22)

What is claimed is:
1. A method of treating a patient having an amyloid deposition disease comprising the step of administering to the patient
a) a therapeutically effective dose of at least one immunoglobulin polypeptide or a fragments thereof, wherein the immunoglobulin polypeptide or fragment thereof binds to an amyloid fibril; and
b) a pharmaceutically acceptable carrier.
2. The method of claim 1, wherein the immunoglobulin polypeptide or fragment thereof is raised against an immunoglobulin light-chain.
3. The method of claim 1, wherein binding of the immunoglobulin polypeptide or fragment thereof opsonizes the amyloid fibril.
4. The method of claim 1, wherein the immunoglobulin polypeptide or fragment thereof is a monoclonal antibody.
5. The method of claim 4, wherein the monoclonal antibody is a humanized antibody.
6. The method of claim 4, wherein the monoclonal antibody is a chimeric antibody.
7. The method of claim 6, wherein the chimeric antibody is a humanized antibody.
8. The method of claim 4, wherein the antibody is a labeled antibody.
9. The method of claim 4, wherein the monoclonal antibody is selected from the group consisting of κ1 (57-18H12), κ4 (11-1F4), λ8 (31-8C7), and combinations thereof.
10. An immunoglobulin polypeptide or fragment thereof that binds to an amyloid fibril and is effective to enhance the cellular immune response of a patient to remove disease-associated amyloid fibril deposits.
11. The immunoglobulin polypeptide or fragment thereof of claim 10, wherein the immunoglobulin polypeptide or fragment thereof is a monoclonal antibody or fragment thereof.
12. The immunoglobulin or fragment thereof of claim 11, wherein the monoclonal antibody is a humanized antibody.
13. The immunoglobulin polypeptide or fragment thereof of claim 11, wherein the monoclonal antibody is a chimeric antibody.
14. The immunoglobulin polypeptide or fragment thereof of claim 13, wherein the chimeric antibody is a humanized antibody.
15. The immunoglobulin polypeptide or fragment thereof of claim 11, wherein the antibody is a labeled antibody.
16. The immunoglobulin polypeptide or fragment thereof of claim 11, wherein the monoclonal antibody is selected from the group consisting of κ1 (57-18H12), κ4 (11-1F4), λ8 (31-8C7), and combinations thereof.
17. The monoclonal antibody or fragment thereof of claim 16, wherein the monoclonal antibody is a humanized antibody.
18. The immunoglobulin polypeptide or fragment thereof of claim 10, wherein the immunoglobulin polypeptide or fragment thereof has been raised against synthetic amyloid fibrils.
19. A pharmaceutical composition comprising the immunoglobulin peptide or fragment thereof of claim 10.
20. A nucleic acid molecule which encodes a polypeptide comprising at least a hypervariable region of the immunoglobulin polypeptide of claim 10.
21. A host cell comprising a nucleic acid molecule of claim 20.
22. A method of producing an immunoglobulin polypeptide comprising the step of culturing the host cell of claim 21.
US09/316,387 1998-05-21 1999-05-21 Methods for amyloid removal using anti-amyloid antibodies Abandoned US20030147882A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/316,387 US20030147882A1 (en) 1998-05-21 1999-05-21 Methods for amyloid removal using anti-amyloid antibodies
US12/693,085 US8105594B2 (en) 1998-05-21 2010-01-25 Methods for amyloid removal using anti-amyloid antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US8619898P 1998-05-21 1998-05-21
US09/316,387 US20030147882A1 (en) 1998-05-21 1999-05-21 Methods for amyloid removal using anti-amyloid antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/693,085 Continuation US8105594B2 (en) 1998-05-21 2010-01-25 Methods for amyloid removal using anti-amyloid antibodies

Publications (1)

Publication Number Publication Date
US20030147882A1 true US20030147882A1 (en) 2003-08-07

Family

ID=27667867

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/316,387 Abandoned US20030147882A1 (en) 1998-05-21 1999-05-21 Methods for amyloid removal using anti-amyloid antibodies
US12/693,085 Expired - Fee Related US8105594B2 (en) 1998-05-21 2010-01-25 Methods for amyloid removal using anti-amyloid antibodies

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/693,085 Expired - Fee Related US8105594B2 (en) 1998-05-21 2010-01-25 Methods for amyloid removal using anti-amyloid antibodies

Country Status (1)

Country Link
US (2) US20030147882A1 (en)

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030165496A1 (en) * 2000-12-06 2003-09-04 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US20040087777A1 (en) * 2000-12-06 2004-05-06 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US20040146512A1 (en) * 2002-10-09 2004-07-29 Arnon Rosenthal Methods of treating Alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
US20040171815A1 (en) * 1997-12-02 2004-09-02 Schenk Dale B. Humanized antibodies that recognize beta amyloid peptide
US20040213800A1 (en) * 2003-02-01 2004-10-28 Seubert Peter A. Active immunization to generate antibodies to soluble A-beta
US20040247590A1 (en) * 2000-05-26 2004-12-09 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050019328A1 (en) * 1997-12-02 2005-01-27 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6913745B1 (en) 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US20050255122A1 (en) * 1997-12-02 2005-11-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20060057701A1 (en) * 2004-07-30 2006-03-16 Arnon Rosenthal Antibodies directed against amyloid-beta peptide and methods using same
US20060252829A1 (en) * 2005-04-15 2006-11-09 Denis Garceau Formulations and methods for treating amyloidosis
US20060292152A1 (en) * 2005-04-29 2006-12-28 Arnon Rosenthal Antibodies directed against amyloid-beta peptide and methods using same
US20080103157A1 (en) * 2006-10-13 2008-05-01 Zenyaku Kogyo Kabushiki Kaisha Methods for treating depression, neurodegeneration, inhibiting amyloid beta deposition, delaying senescence, and extending life spans with heterocyclic compounds
US20080248029A1 (en) * 1999-06-01 2008-10-09 Schenk Dale B Prevention and treatment of amyloidogenic diseases
US20090017040A1 (en) * 2007-06-12 2009-01-15 Ac Immune S.A. Monoclonal antibody
US20090017041A1 (en) * 2007-06-12 2009-01-15 Ac Immune S.A. Monoclonal antibody
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US7772375B2 (en) 2005-12-12 2010-08-10 Ac Immune S.A. Monoclonal antibodies that recognize epitopes of amyloid-beta
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20100297012A1 (en) * 2007-10-05 2010-11-25 Andrea Pfeifer Humanized antibody
US20100322932A1 (en) * 1998-05-21 2010-12-23 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7892544B2 (en) 2006-07-14 2011-02-22 Ac Immune Sa Humanized anti-beta-amyloid antibody
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US20110212109A1 (en) * 2006-11-30 2011-09-01 Stefan Barghorn Abeta CONFORMER SELECTIVE ANTI-Abeta GLOBULOMER MONOCLONAL ANTIBODIES
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US8987419B2 (en) 2010-04-15 2015-03-24 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US9221900B2 (en) 2010-07-30 2015-12-29 Ac Immune S.A. Methods for identifying safe and functional humanized antibodies
US9403902B2 (en) 2007-10-05 2016-08-02 Ac Immune S.A. Methods of treating ocular disease associated with amyloid-beta-related pathology using an anti-amyloid-beta antibody
US9540432B2 (en) 2005-11-30 2017-01-10 AbbVie Deutschland GmbH & Co. KG Anti-Aβ globulomer 7C6 antibodies
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090232733A1 (en) * 2005-04-13 2009-09-17 O'nuallain Brian Diagnostic and Therapeutic Potential of Immune Globulin Intravenous (IGIV) Products
CA3067597C (en) 2017-06-29 2023-03-21 The Trustees Of Columbia University In The City Of New York Chimeric antibodies for treatment of amyloid deposition diseases
US11382974B2 (en) * 2017-08-01 2022-07-12 The Trustees Of Columbia University In The City Of New York Methods and compositions for treatment of amyloid deposition diseases
WO2020167376A1 (en) 2019-02-12 2020-08-20 Protheima Biosciences Limited Treatment of al amyloidosis with the combination of monoclonal antibodies against immunoglobulin light chains and the cd38 cell membrane molecule on antibody-producing and other immune cells
AU2020231081A1 (en) 2019-03-05 2021-09-30 Prothena Biosciences Limited Methods of treating AL amyloidosis
AU2021402185A1 (en) * 2020-12-14 2023-07-20 University Health Network Immunoglobulin light chain antibodies and uses thereof
TW202300517A (en) 2021-03-12 2023-01-01 美商美國禮來大藥廠 Anti-amyloid beta antibodies and uses thereof
WO2022246433A1 (en) * 2021-05-18 2022-11-24 University Of Tennessee Research Foundation Antibody-peptide fusion proteins for treating amyloid disorders
WO2022251048A1 (en) 2021-05-24 2022-12-01 Eli Lilly And Company Anti-amyloid beta antibodies and uses thereof

Family Cites Families (158)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CU22545A1 (en) 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
US5208036A (en) 1985-01-07 1993-05-04 Syntex (U.S.A.) Inc. N-(ω, (ω-1)-dialkyloxy)- and N-(ω, (ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4666829A (en) 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
US4713366A (en) 1985-12-04 1987-12-15 The Ohio State University Research Foundation Antigenic modification of polypeptides
US5231170A (en) 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
US5220013A (en) 1986-11-17 1993-06-15 Scios Nova Inc. DNA sequence useful for the detection of Alzheimer's disease
US5187153A (en) 1986-11-17 1993-02-16 Scios Nova Inc. Methods of treatment using Alzheimer's amyloid polypeptide derivatives
DE3702789A1 (en) 1987-01-30 1988-08-18 Bayer Ag PROCUREMENT PROTEIN OF APC POLYPEPTIDE, FOR CODING DNA AND DIAGNOSTIC USE OF DNA AND PROTEIN
JPS63245689A (en) 1987-03-31 1988-10-12 Suntory Ltd Human amyloid-related protein monoclonal antibody
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5583112A (en) 1987-05-29 1996-12-10 Cambridge Biotech Corporation Saponin-antigen conjugates and the use thereof
US5869054A (en) 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of autoantigens
US5571500A (en) 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases through administration by inhalation of autoantigens
US5571499A (en) 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5849298A (en) 1987-06-24 1998-12-15 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of bovine myelin
JP2512796B2 (en) 1987-06-24 1996-07-03 オートイミュン・インコーポレイテッド Treatment of autoimmune disease by oral administration of self-antigen
US5641474A (en) 1987-06-24 1997-06-24 Autoimmune, Inc. Prevention of autoimmune diseases by aerosol administration of autoantigens
US5645820A (en) 1987-06-24 1997-07-08 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5004697A (en) 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
CA1339014C (en) 1987-10-08 1997-03-25 Ronald E. Majocha Antibodies to a4 amyloid peptide
US5231000A (en) 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
WO1989003687A1 (en) 1987-10-23 1989-05-05 Genetics Institute, Inc. Composition and method for treating cancers characterized by over-expression of the c-fms proto-oncogene
WO1989006689A1 (en) 1988-01-13 1989-07-27 The Mclean Hospital Corporation Genetic constructs containing the alzheimer brain amyloid gene
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
WO1990012870A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
WO1990012871A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
ES2144398T3 (en) 1989-12-20 2000-06-16 Autoimmune Inc IMPROVED TREATMENT OF AUTOIMMUNE DISEASES THROUGH THE ADMINISTRATION IN SELF-ANTIGEN SPRAY.
JPH05508621A (en) 1990-03-02 1993-12-02 オートイミューン インク Down control of autoimmune diseases by oral administration of autoantigens
EP0451700A1 (en) 1990-04-10 1991-10-16 Miles Inc. Recombinant APP minigenes for expression in transgenic mice as models for Alzheimers's disease
CA2079880A1 (en) 1990-04-24 1991-10-25 William E. Van Nostrand Purification, detection and methods of use of protease nexin-2
US5753624A (en) 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
ATE153534T1 (en) 1990-04-27 1997-06-15 John Mcmichael METHOD AND COMPOSITION FOR TREATING CNS DISEASES CAUSED BY ABNORMAL BETA-AMYLOID PROTEIN
ES2217250T3 (en) 1990-06-15 2004-11-01 Scios Inc. TRANSGENIC, NON-HUMAN MAMMER THAT SHOWS THE AMILOID TRAINING PATHOLOGY OF ALZHEIMER'S DISEASE.
US5780587A (en) 1990-08-24 1998-07-14 President And Fellows Of Harvard College Compounds and methods for inhibiting β-protein filament formation and neurotoxicity
JPH06502071A (en) 1990-09-28 1994-03-10 ジ・アップジョン・カンパニー Transgenic animals carrying Alzheimer's amyloid precursor gene
KR0140841B1 (en) 1990-10-15 1998-06-01 조앤 월리스 Treatment of autoimmune diseases by oral administration of autoantigens
NZ241311A (en) 1991-01-17 1995-03-28 Gen Hospital Corp Rna sequence having trans-splicing activity, plant strains
ATE286971T1 (en) 1991-01-21 2005-01-15 Elan Pharm Inc TEST AND MODEL FOR ALZHEIMERS DISEASE
FI90464C (en) 1991-02-21 1994-02-10 Kemira Oy Catalyst
US5192753A (en) 1991-04-23 1993-03-09 Mcgeer Patrick L Anti-rheumatoid arthritic drugs in the treatment of dementia
US5672805A (en) 1991-07-18 1997-09-30 The Regents Of The University Of California Transgenic mice expressing the neurotoxic C-terminus of β-amyloid precursor protein
US5434050A (en) 1991-08-13 1995-07-18 Regents Of The University Of Minnesota Labelled β-amyloid peptide and methods of screening for Alzheimer's disease
AU671093B2 (en) 1992-01-07 1996-08-15 Elan Pharmaceuticals, Inc. Transgenic animal models for alzheimer's disease
US5679348A (en) 1992-02-03 1997-10-21 Cedars-Sinai Medical Center Immunotherapy for recurrent HSV infections
KR950700081A (en) 1992-02-11 1995-01-16 W 로우 죤 Dual Carrier Immunogenic Constructs
KR950700082A (en) 1992-02-28 1995-01-16 로버트 씨. 비숍 How to Treat Autoimmune Diseases by Bystander
EP0561087B1 (en) 1992-03-20 1999-08-04 N.V. Innogenetics S.A. Mutated form of the beta-amyloid precursor protein gene
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
US5441870A (en) 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
GB9209118D0 (en) 1992-04-28 1992-06-10 Sb 120 Amsterdam Bv Vaccine compositions
US5837672A (en) 1992-07-10 1998-11-17 Athena Neurosciences, Inc. Methods and compositions for the detection of soluble β-amyloid peptide
US5766846A (en) 1992-07-10 1998-06-16 Athena Neurosciences Methods of screening for compounds which inhibit soluble β-amyloid peptide production
WO1994001772A1 (en) 1992-07-13 1994-01-20 The Children's Medical Center Corporation SCREEN FOR ALZHEIMER'S DISEASE THERAPEUTICS BASED ON β-AMYLOID PRODUCTION
ES2127829T3 (en) 1992-07-31 1999-05-01 Medeva Holdings Bv EXPRESSION OF RECOMBINANT PROTEINS FUSED IN ATTENUATED BACTERIA.
GB9216360D0 (en) 1992-07-31 1992-09-16 Health Care South West Durham Treatment of blood
US5958883A (en) 1992-09-23 1999-09-28 Board Of Regents Of The University Of Washington Office Of Technology Animal models of human amyloidoses
US5605811A (en) 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5750349A (en) 1993-01-25 1998-05-12 Takeda Chemical Industries Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5955317A (en) 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
CA2115900A1 (en) 1993-02-22 1994-08-23 Gerald W. Becker Pharmaceutical screens and antibodies
PT812593E (en) 1993-03-23 2002-01-30 Smithkline Beecham Biolog VACCINES CONTAINING 3-O-DEACILED MONOPHOSPHALOR-LIPID IN ITS COMPOSITION
DK1060750T3 (en) 1993-03-29 2006-01-09 Univ Kingston Propane-1,3-disulfonic acid and its pharmaceutically acceptable salts for the treatment of amyloidosis
AU7043894A (en) 1993-05-28 1994-12-20 Miriam Hospital, The Composition and method for (in vivo) imaging of amyloid deposits
US5464823A (en) 1993-07-20 1995-11-07 The Regents Of The University Of California Mammalian antibiotic peptides
JPH09500540A (en) 1993-07-30 1997-01-21 メデヴァ ホールディングス ビー.ヴイ. Vaccine composition
GB9317120D0 (en) 1993-08-17 1993-09-29 Royal Postgrad Med School Human serum amyloid p component
WO1995005853A1 (en) 1993-08-26 1995-03-02 The Regents Of The University Of California Method, compositions and devices for administration of naked polynucleotides which encode biologically active peptides
AU698962B2 (en) 1993-09-14 1998-11-12 Epimmune, Inc. Alteration of immune response using pan DR-binding peptides
ATE213507T1 (en) 1993-10-20 2002-03-15 Univ Duke METHOD FOR BINDING MATERIAL TO BETA-AMYLOID PEPTIDE
CA2172507C (en) 1993-10-22 2008-12-02 Jeffrey L. Cleland Methods and compositions for microencapsulation of antigens for use as vaccines
US5744368A (en) 1993-11-04 1998-04-28 Research Foundation Of State University Of New York Methods for the detection of soluble amyloid β-protein (βAP) or soluble transthyretin (TTR)
GB9326253D0 (en) 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
US5434170A (en) 1993-12-23 1995-07-18 Andrulis Pharmaceuticals Corp. Method for treating neurocognitive disorders
US5877399A (en) 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
CA2182311A1 (en) 1994-01-27 1995-08-03 Karen Hsiao Transgenic non-human mammals with progressive neurologic disease
CA2182731A1 (en) 1994-02-03 1995-08-10 Michael P. Vitek Compositions and methods for advanced glycosylation endproduct-mediated modulation of amyloidosis
ATE202940T1 (en) 1994-05-25 2001-07-15 John Mcmichael MEANS AND METHODS FOR TREATING PLAQUE DISEASES
US5622701A (en) 1994-06-14 1997-04-22 Protein Design Labs, Inc. Cross-reacting monoclonal antibodies specific for E- and P-selectin
US6417178B1 (en) 1994-07-19 2002-07-09 University Of Pittsburgh Amyloid binding nitrogen-linked compounds for the antemortem diagnosis of alzheimer's disease, in vivo imaging and prevention of amyloid deposits
US6114133A (en) 1994-11-14 2000-09-05 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of Alzheimer's disease by measuring amyloid-β peptide (x-≧41)
US5688651A (en) 1994-12-16 1997-11-18 Ramot University Authority For Applied Research And Development Ltd. Prevention of protein aggregation
US5786180A (en) 1995-02-14 1998-07-28 Bayer Corporation Monoclonal antibody 369.2B specific for β A4 peptide
ES2175083T3 (en) 1995-03-14 2002-11-16 Praecis Pharm Inc AMULOID AGGREGATION MODULATORS.
WO1996039176A1 (en) 1995-06-05 1996-12-12 Brigham & Women's Hospital USE OF ORAL TOLERANCE TO SUPPRESS BOTH Th1 AND Th2 IMMUNE RESPONSES AND TO SUPPRESS ANTIBODY PRODUCTION
ES2174027T3 (en) 1995-06-30 2002-11-01 American Cyanamid Co STABLE COMPOSITIONS CONTAINING MACROLIDES AND MACROLIDES COMBINED WITH VACCINES.
AU6898996A (en) 1995-08-21 1997-03-12 Cytrx Corporation Compositions and methods for growth promotion
ATE263374T1 (en) 1995-09-14 2004-04-15 Univ California ANTIBODIES SPECIFIC TO NATIVE PRP-SC
US5750361A (en) 1995-11-02 1998-05-12 The Regents Of The University Of California Formation and use of prion protein (PRP) complexes
PT859959E (en) 1995-11-10 2003-12-31 Elan Corp Plc PEPTIDES THAT INCREASE TRANSPORTATION THROUGH TISSUES AND METHODS FOR THEIR IDENTIFICATION AND USES
WO1997021728A1 (en) 1995-12-12 1997-06-19 Karolinska Innovations Ab PEPTIDE BINDING THE KLVFF-SEQUENCE OF AMYLOID $g(b)
JPH09178743A (en) 1995-12-27 1997-07-11 Oriental Yeast Co Ltd Determinationof soluble app
US6150091A (en) 1996-03-06 2000-11-21 Baylor College Of Medicine Direct molecular diagnosis of Friedreich ataxia
JP4229341B2 (en) 1996-03-23 2009-02-25 財団法人阪大微生物病研究会 Tetanus toxin functional fragment antigen and tetanus vaccine
US6284533B1 (en) 1996-05-01 2001-09-04 Avant Immunotherapeutics, Inc. Plasmid-based vaccine for treating atherosclerosis
CA2254931C (en) 1996-06-11 2007-04-10 Yeda Research And Development Co. Ltd. Human monoclonal antibodies to the hepatitis b surface antigen
CA2183901A1 (en) 1996-08-22 1998-02-23 Johanna E. Bergmann Targets for therapy and diagnosis of alzheimer's disease and down syndrome in humans
US6057367A (en) 1996-08-30 2000-05-02 Duke University Manipulating nitrosative stress to kill pathologic microbes, pathologic helminths and pathologically proliferating cells or to upregulate nitrosative stress defenses
US6797495B2 (en) 1996-11-05 2004-09-28 The Regents Of The University Of California Somatic cells with ablated PrP gene and methods of use
US20020086847A1 (en) 1997-04-09 2002-07-04 Mindset Biopharmaceuticals (Usa) Recombinant antibodies specific for beta-amyloid ends, DNA encoding and methods of use thereof
SI0994728T1 (en) 1997-04-09 2009-02-28 Intellect Neurosciences Inc Recombinant antibodies specific for beta-amyloid ends, dna encoding and methods of use thereof
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
AU8269898A (en) 1997-06-27 1999-01-19 Regents Of The University Of California, The Drug targeting of a peptide radiopharmaceutical through the primate blood-brain barrier in vivo with a monoclonal antibody to the human insulin receptor
IT1293511B1 (en) 1997-07-30 1999-03-01 Gentili Ist Spa MONOCLONAL CATALYTIC ANTIBODIES WITH PROTEASIC ACTIVITY FOR THE SELECTIVE LYSIS OF THE PROTEIN COMPONENT OF PLATES AND RELATED AGGREGATES
WO1999006545A2 (en) 1997-08-01 1999-02-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Composition and method for the detection of diseases associated with amyloid-like fibril or protein aggregate formation
EP0911035A3 (en) 1997-10-24 2002-08-21 Eli Lilly And Company Insoluble insulin compositions
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6743427B1 (en) 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6761888B1 (en) 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
DE69831971T2 (en) 1997-12-03 2006-07-06 Neuralab Ltd., Flatts SUPPRESSION OF CHANGES ASSOCIATED WITH BETA AMYLOID AT ALZHEIMER
BR9815345A (en) 1997-12-03 2000-11-21 Fujisawa Pharmaceutical Co Soft composition of pelleted drug, inhaler using it and method for its manufacture
FR2777015B3 (en) 1998-02-23 2000-09-15 Financ De Biotechnologie METHOD AND MEANS FOR OBTAINING CELLULAR AND ANIMAL MODELS OF NEURODEGENERATIVE DISEASES
NO314086B1 (en) 1998-05-08 2003-01-27 Gemvax As Peptides and pharmaceutical compositions containing them, nucleic acid sequences encoding such peptides, plasmids and virus vectors encompassing such DNA sequences and their use for the preparation of pharmaceutical preparations for
JPH11322788A (en) 1998-05-08 1999-11-24 Ajinomoto Co Inc New aspartame derivative crystal and its production
JP2003532618A (en) 1998-05-19 2003-11-05 イエダ リサーチ アンド デベロプメント カンパニイ リミテッド Cells, nervous system-specific antigens and their uses
JP2002515235A (en) 1998-05-21 2002-05-28 ザ ユニヴァーシティ オブ テネシー リサーチ コーポレイション Amyloid removal method using anti-amyloid antibody
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US6170226B1 (en) 1998-07-15 2001-01-09 William Chang Capsule filling apparatus
EP1148891B1 (en) 1999-01-19 2004-03-17 PHARMACIA & UPJOHN COMPANY Method of packaging an oxidation sensitive medicinal substance
JP2002535289A (en) 1999-01-22 2002-10-22 ドウアリング,マシユー・ジヨン Vaccine-mediated treatment of neurological disorders
US6838592B1 (en) 1999-04-30 2005-01-04 Nathan S. Kline Institute For Psychiatric Research Methods for the identification of compounds for the treatment of alzheimer's disease
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
NO311648B1 (en) 1999-05-31 2001-12-27 Genomar Asa DNA sequences that can be used for vaccine, preventive health work on fish and aquatic organisms and otherwise within biomedicine, vector, ILA virus vaccine and diagnostic set
AR024558A1 (en) 1999-06-01 2002-10-16 Neuralab Ltd COMPOSITIONS OF THE A-BETA PEPTIDE AND PROCESSES TO PRODUCE THE SAME
US20040146521A1 (en) 1999-06-01 2004-07-29 Schenk Dale B. Prevention and treatment of synucleinopathic disease
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
DE60040024D1 (en) 1999-06-16 2008-10-02 Boston Biomedical Res Inst IMMUNOLOGICAL CONTROL OF THE BETA-AMYLOID CONTENT IN VIVO
US6294171B2 (en) 1999-09-14 2001-09-25 Milkhaus Laboratory, Inc. Methods for treating disease states comprising administration of low levels of antibodies
US20020094335A1 (en) 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
AU784312B2 (en) 1999-11-29 2006-03-09 Bellus Health (International) Limited Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
US20030049251A1 (en) 1999-12-08 2003-03-13 Barbas Carlos F. Methods and compositions useful for inhibiting ccr5-dependent infection of cells by hiv-1
ES2275570T3 (en) 1999-12-08 2007-06-16 Intellect Neurosciences, Inc. BETA CHEMERIC AMYLOID PEPTIDES.
US6399314B1 (en) 1999-12-29 2002-06-04 American Cyanamid Company Methods of detection of amyloidogenic proteins
CN1279971C (en) 2000-02-21 2006-10-18 法麦克萨有限公司 Novel method for down-regulation of amyloid
AU783144B2 (en) 2000-02-21 2005-09-29 H. Lundbeck A/S Novel method for down-regulation of amyloid
IT1320890B1 (en) 2000-02-22 2003-12-10 Massimiliano Sichera DEVICE FOR THE INSERTION OF ITEMS WITHIN THE RELATIVE CONTAINERS
AU2001241636A1 (en) 2000-02-23 2001-09-03 The Procter And Gamble Company Halotherapy method
SK288723B6 (en) 2000-02-24 2020-01-07 Univ Washington Pharmaceutical preparation and use of said pharmaceutical preparation
AU2001253158A1 (en) 2000-04-05 2001-10-23 University Of Tennessee Research Corporation Methods of investigating, diagnosing, and treating amyloidosis
ATE286072T1 (en) 2000-05-22 2005-01-15 Univ New York SYNTHETIC IMMUNOGENIC BUT NON-AMYLOIDOGENIC PEPTIDES HOMOLOGUE TO AMYLOID BETA AND THEIR USE TO INDUCE AN IMMUNE RESPONSE AGAINST AMYLOID BETA AND AMYLOID AGGREGATES
WO2002003911A2 (en) 2000-07-07 2002-01-17 Lars Lannfelt Prevention and treatment of alzheimer's disease
EP1172378A1 (en) 2000-07-12 2002-01-16 Richard Dr. Dodel Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
US20020009445A1 (en) 2000-07-12 2002-01-24 Yansheng Du Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
IT1319277B1 (en) 2000-10-24 2003-09-26 Chiesi Farma Spa MELTING PROTEINS USEFUL FOR ALZHEIMER'S MILK IMMUNIZATION TREATMENT.
IL139308A0 (en) 2000-10-26 2001-11-25 Marikovsky Moshe Peptides from amyloid precursor protein which inhibit tumor growth and metastasis
PT1346041E (en) 2000-11-27 2007-06-05 Praecis Pharm Inc Therapeutic agents and methods of use thereof for treating an amyloidogenic disease
PE20020574A1 (en) 2000-12-06 2002-07-02 Wyeth Corp HUMANIZED ANTIBODIES THAT RECOGNIZE THE AMYLOID PEPTIDE BETA
US6900036B2 (en) 2000-12-27 2005-05-31 University Of Texas Health Science Center Houston Prion isomers, methods of making, methods of using, and compositions and products comprising prion isomers
WO2002059621A2 (en) 2001-01-24 2002-08-01 Bayer Corporation Regulation of transthyretin to treat obesity
DE60121729T2 (en) 2001-04-19 2007-11-29 Dr. Hermann Schätzl Prion protein dimers for vaccinations
US6906169B2 (en) 2001-05-25 2005-06-14 United Biomedical, Inc. Immunogenic peptide composition comprising measles virus Fprotein Thelper cell epitope (MUFThl-16) and N-terminus of β-amyloid peptide
US20020197258A1 (en) 2001-06-22 2002-12-26 Ghanbari Hossein A. Compositions and methods for preventing protein aggregation in neurodegenerative diseases
CA2407163C (en) 2002-10-09 2010-12-14 Stackteck Systems Ltd. Valve pin cross over nozzle
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US20090232733A1 (en) 2005-04-13 2009-09-17 O'nuallain Brian Diagnostic and Therapeutic Potential of Immune Globulin Intravenous (IGIV) Products

Cited By (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196399A1 (en) * 1997-12-02 2005-09-08 Schenk Dale B. Prevention and treatment of amyloidogenic disease
US8642044B2 (en) 1997-12-02 2014-02-04 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20040171815A1 (en) * 1997-12-02 2004-09-02 Schenk Dale B. Humanized antibodies that recognize beta amyloid peptide
US20040171816A1 (en) * 1997-12-02 2004-09-02 Schenk Dale B. Humanized antibodies that recognize beta amyloid peptide
US6946135B2 (en) 1997-12-02 2005-09-20 Neuralab Limited Prevention and treatment of amyloidogenic disease
US8034348B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US20110306756A1 (en) * 1997-12-02 2011-12-15 Janssen Alzheimer Immunotherapy Prevention and Treatment of Amyloidogenic Disease
US20050019328A1 (en) * 1997-12-02 2005-01-27 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050019343A1 (en) * 1997-12-02 2005-01-27 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050037026A1 (en) * 1997-12-02 2005-02-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US20050142132A1 (en) * 1997-12-02 2005-06-30 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6913745B1 (en) 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US20050163788A1 (en) * 1997-12-02 2005-07-28 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050191314A1 (en) * 1997-12-02 2005-09-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US20050249725A1 (en) * 1997-12-02 2005-11-10 Schenk Dale B Humanized antibodies that recognize beta amyloid peptide
US6972127B2 (en) 1997-12-02 2005-12-06 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050255122A1 (en) * 1997-12-02 2005-11-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US8105594B2 (en) 1998-05-21 2012-01-31 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20100322932A1 (en) * 1998-05-21 2010-12-23 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20090285809A1 (en) * 1999-06-01 2009-11-19 Elan Pharmaceuticals, Inc. Prevention and treatment of amyloidogenic diseases
US20110182893A1 (en) * 1999-06-01 2011-07-28 Elan Pharma International Limited Prevention and treatment of amyloidogenic diseases
US20080248029A1 (en) * 1999-06-01 2008-10-09 Schenk Dale B Prevention and treatment of amyloidogenic diseases
US20110177066A1 (en) * 1999-06-01 2011-07-21 Elan Pharma International Limited Prevention and treatment of amyloidogenic diseases
US8124081B2 (en) * 1999-06-01 2012-02-28 Crimagua Limited Prevention and treatment of amyloidogenic diseases
US20090285822A1 (en) * 1999-06-01 2009-11-19 Elan Pharmaceuticals, Inc. Prevention and treatment of amyloidogenic disease
US7977316B2 (en) 1999-06-01 2011-07-12 Elan Pharmaceuticals, Inc. Prevention and treatment of amyloidogenic diseases
US20040265301A1 (en) * 2000-05-26 2004-12-30 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20040247591A1 (en) * 2000-05-26 2004-12-09 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050158304A1 (en) * 2000-05-26 2005-07-21 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20040247590A1 (en) * 2000-05-26 2004-12-09 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20060121038A9 (en) * 2000-05-26 2006-06-08 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050123544A1 (en) * 2000-05-26 2005-06-09 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US20030165496A1 (en) * 2000-12-06 2003-09-04 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US20060280743A1 (en) * 2000-12-06 2006-12-14 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US20040087777A1 (en) * 2000-12-06 2004-05-06 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
US20070160616A1 (en) * 2002-10-09 2007-07-12 Arnon Rosenthal Methods of treating Alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
US20040146512A1 (en) * 2002-10-09 2004-07-29 Arnon Rosenthal Methods of treating Alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US20040213800A1 (en) * 2003-02-01 2004-10-28 Seubert Peter A. Active immunization to generate antibodies to soluble A-beta
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7927594B2 (en) 2004-07-30 2011-04-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide
US7807165B2 (en) 2004-07-30 2010-10-05 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US20060057701A1 (en) * 2004-07-30 2006-03-16 Arnon Rosenthal Antibodies directed against amyloid-beta peptide and methods using same
US20110008834A1 (en) * 2004-07-30 2011-01-13 Rinat Neuroscience Corp. Polynucleotides encoding antibodies directed against amyloid-beta peptide
US8268593B2 (en) 2004-07-30 2012-09-18 Rinat Neuroscience Corp. Polynucleotides encoding antibodies directed against amyloid-beta peptide
US20060057702A1 (en) * 2004-07-30 2006-03-16 Arnon Rosenthal Antibodies directed against amyloid-beta peptide and methods using same
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US20060252829A1 (en) * 2005-04-15 2006-11-09 Denis Garceau Formulations and methods for treating amyloidosis
US8178580B2 (en) * 2005-04-15 2012-05-15 Kiacta Sarl Formulations and methods for treating amyloidosis
US7763250B2 (en) 2005-04-29 2010-07-27 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and nucleic acids encoding same
US8398978B2 (en) 2005-04-29 2013-03-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US20060292152A1 (en) * 2005-04-29 2006-12-28 Arnon Rosenthal Antibodies directed against amyloid-beta peptide and methods using same
US9540432B2 (en) 2005-11-30 2017-01-10 AbbVie Deutschland GmbH & Co. KG Anti-Aβ globulomer 7C6 antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US7772375B2 (en) 2005-12-12 2010-08-10 Ac Immune S.A. Monoclonal antibodies that recognize epitopes of amyloid-beta
US20110070613A1 (en) * 2005-12-12 2011-03-24 Ac Immune S.A. Monoclonal Antibody
US20100297132A1 (en) * 2005-12-12 2010-11-25 Ac Immune S.A. Monoclonal antibody
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US7892544B2 (en) 2006-07-14 2011-02-22 Ac Immune Sa Humanized anti-beta-amyloid antibody
US8246954B2 (en) 2006-07-14 2012-08-21 Ac Immune S.A. Methods of treating amyloidosis with humanized anti-beta-amyloid antibodies
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
US8124353B2 (en) 2006-07-14 2012-02-28 Ac Immune S.A. Methods of treating and monitoring disease with antibodies
US9089561B2 (en) 2006-10-13 2015-07-28 Zenyaku Kogyo Kabushiki Kaisha Methods for treating depression, neurodegeneration, inhibiting amyloid β deposition, delaying senescence, and extending life spans with heterocyclic compounds
US20080103157A1 (en) * 2006-10-13 2008-05-01 Zenyaku Kogyo Kabushiki Kaisha Methods for treating depression, neurodegeneration, inhibiting amyloid beta deposition, delaying senescence, and extending life spans with heterocyclic compounds
US20110212109A1 (en) * 2006-11-30 2011-09-01 Stefan Barghorn Abeta CONFORMER SELECTIVE ANTI-Abeta GLOBULOMER MONOCLONAL ANTIBODIES
US8877190B2 (en) 2006-11-30 2014-11-04 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9359430B2 (en) 2006-11-30 2016-06-07 Abbvie Inc. Abeta conformer selective anti-Abeta globulomer monoclonal antibodies
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US9146244B2 (en) 2007-06-12 2015-09-29 Ac Immune S.A. Polynucleotides encoding an anti-beta-amyloid monoclonal antibody
US9585956B2 (en) 2007-06-12 2017-03-07 Ac Immune S.A. Polynucleotides encoding anti-amyloid beta monoclonal antibodies
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
US20090017041A1 (en) * 2007-06-12 2009-01-15 Ac Immune S.A. Monoclonal antibody
US20090017040A1 (en) * 2007-06-12 2009-01-15 Ac Immune S.A. Monoclonal antibody
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US9403902B2 (en) 2007-10-05 2016-08-02 Ac Immune S.A. Methods of treating ocular disease associated with amyloid-beta-related pathology using an anti-amyloid-beta antibody
US20100297012A1 (en) * 2007-10-05 2010-11-25 Andrea Pfeifer Humanized antibody
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US8987419B2 (en) 2010-04-15 2015-03-24 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9221900B2 (en) 2010-07-30 2015-12-29 Ac Immune S.A. Methods for identifying safe and functional humanized antibodies
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins

Also Published As

Publication number Publication date
US20100322932A1 (en) 2010-12-23
US8105594B2 (en) 2012-01-31

Similar Documents

Publication Publication Date Title
US8105594B2 (en) Methods for amyloid removal using anti-amyloid antibodies
EP1078005B1 (en) Methods for amyloid removal using anti-amyloid antibodies
TWI705975B (en) ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
KR100767146B1 (en) Humanized Antibodies That Sequester Amyloid Beta Peptide
EP0805871B2 (en) Anti-cd30 antibodies preventing proteolytic cleavage and release of membrane-bound cd30 antigen
JP2020147574A (en) Anti-age antibodies for treating inflammation and auto-immune disorders
US20060025575A1 (en) Immunological detection of prions
JPH10505819A (en) Use of autoantibodies for treatment and prevention of tumors
KR20160093726A (en) Antibody-based therapy of transthyretin(ttr) amyloidosis and human-derived antibodies therefor
JP2022548937A (en) ANTI-ALPHA-HEMOLYSIN ANTIBODY AND USES THEREOF
WO2007119447A1 (en) Therapeutic agent for rheumatoid arthritis
JPH05503687A (en) Treatment of autoimmune diseases
CN106831983A (en) For the humanized antibody of amyloid-beta
AU2003262458B2 (en) Methods for amyloid removal using anti-amyloid antibodies
JP4044733B2 (en) Humanized antibody recognizing verotoxin II and cell line producing the antibody
US20030180799A1 (en) Antibodies against plasma cells
JP2004532626A (en) Polyclonal populations of bispecific molecules, and methods for their production and use
CN116333145B (en) Antibodies that bind activated factor IX
OA20246A (en) Monoclonal antibodies against the beta chain region of human TRBV9.
OA20245A (en) Monoclonal antibodies that bind specifically to human TRBV9.
WO2023287941A2 (en) Anti-tax interacting protein-1 (tip1) binding molecules
JP2022132961A (en) Vector for gene therapy of rheumatoid arthritis
WO2013027658A1 (en) Cartilage/bone destruction suppressor
KR20160084177A (en) Anti-Mutant Luterial Antibody and Method for Preparing the Same
Ayana The role of monoclonal antibodies in therapy and vaccine developments

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SOLOMON, ALAN;HRNCIC, RUDI;WALL, JONATHAN STUART;REEL/FRAME:010209/0551;SIGNING DATES FROM 19990625 TO 19990709

AS Assignment

Owner name: UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE,

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE'S ADDRESS PREVIOUSLY RECORDED ON REEL 010114, FRAME 0739;ASSIGNORS:SOLOMON, ALAN;HRNCIC, RUDI;WALL, JONATHAN STUART;REEL/FRAME:011412/0727;SIGNING DATES FROM 19990625 TO 19990709

AS Assignment

Owner name: UNIVERSITY OF TENNESSEE RESEARCH FOUNDATION, TENNE

Free format text: CHANGE OF NAME;ASSIGNOR:UNIVERSITY OF TENNESSEE RESEARCH CORPORATION, THE;REEL/FRAME:014733/0805

Effective date: 20030514

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF TENNESSEE;REEL/FRAME:021766/0425

Effective date: 20030508

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION