WO2022093195A1 - Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens - Google Patents

Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens Download PDF

Info

Publication number
WO2022093195A1
WO2022093195A1 PCT/US2020/057539 US2020057539W WO2022093195A1 WO 2022093195 A1 WO2022093195 A1 WO 2022093195A1 US 2020057539 W US2020057539 W US 2020057539W WO 2022093195 A1 WO2022093195 A1 WO 2022093195A1
Authority
WO
WIPO (PCT)
Prior art keywords
age
seq
antibody
composition
sequence identity
Prior art date
Application number
PCT/US2020/057539
Other languages
French (fr)
Inventor
Lewis S. Gruber
Original Assignee
Siwa Corporation
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 Siwa Corporation filed Critical Siwa Corporation
Priority to PCT/US2020/057539 priority Critical patent/WO2022093195A1/en
Publication of WO2022093195A1 publication Critical patent/WO2022093195A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • Osteoarthritis is the general term for the more than 100 different diseases and disorders that affect joints or articulations of the body. Osteoarthritis is the most common type of arthritis and affects approximately 30 million adults in the United States (U.S. Centers for Disease Control and Prevention). Worldwide, it is estimated that 10% of all individuals over age 60 have significant clinical problems that can be attributed to osteoarthritis (World Health Organization).
  • Osteoarthritis results from the deterioration of joint cartilage, which is composed of specialized cells known as chondrocytes. Chondrocytes produce a collagenous extracellular matrix containing collagen and proteoglycans. The loss or breakdown of cartilage increases friction between the bones of the joint, which can lead to the growth of bone spurs (osteophytes) or the release of cartilage and bone fragments into the joint space. Symptoms of osteoarthritis include joint pain, stiffness, reduced range of motion, joint swelling and weakness or numbness of the arms and legs. Osteoarthritis may develop in any joint, but is most commonly seen in the hands and fingers, knees, hips, feet, neck and back.
  • osteoarthritis-associated pain is often treated with over-the-counter analgesics, such as acetaminophen and nonsteroidal anti-inflammatory drugs (NSAIDs), or prescription pain medicines, such as opiates and COX-2 inhibitors.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • Local treatment of an affected joint may involve intra-articular injection of corticosteroids, hyaluronic acid or hyaluronic acid substitutes. In more severe cases, surgery may be required to repair or replace the affected joint.
  • Senescent cells are cells that are partially-functional or non-functional and are in a state of proliferative arrest. Senescence is a distinct state of a cell, and is associated with biomarkers, such as activation of the biomarker p16 lnk4a , and expression of p-galactosidase. Senescence begins with damage or stress (such as overstimulation by growth factors) of cells.
  • AGEs Advanced glycation end-products
  • AGE-modified proteins or glycation end-products
  • AGE-modified proteins or glycation end-products
  • AGEs may also be formed from other processes.
  • the advanced glycation end product, N e -(carboxymethyl)lysine is a product of both lipid peroxidation and glycoxidation reactions.
  • AGEs have been associated with several pathological conditions including diabetic complications, inflammation, retinopathy, nephropathy, atherosclerosis, stroke, endothelial cell dysfunction, and neurodegenerative disorders (Bierhaus A, “AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept,” Cardiovasc Res, Vol. 37(3), 586-600 (1998)).
  • AGE-modified proteins are also a marker of senescent cells. This association between glycation end-product and senescence is well known in the art. See, for example, Gruber, L. (WO 2009/143411, 26 Nov. 2009), Ando, K. et al. (Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation, Biochem Biophys Res Common., Vol. 258, 123, 125 (1999)), Ahmed, E.K. etal. (“Protein Modification and Replicative Senescence of Wl- 38 Human Embryonic Fibroblasts” Aging Cells, vol. 9, 252, 260 (2010)), Vlassara, H.
  • glycation end-products are “one of the major causes of spontaneous damage to cellular and extracellular proteins" (Ahmed, E.K. et al., see above, page 353). Accordingly, the accumulation of glycation endproducts is associated with senescence and lack of function.
  • MG methyl glyoxal
  • Damage or stress to mitochondrial DNA also sets off a DNA damage response which induces the cell to produce cell cycle blocking proteins. These blocking proteins prevent the cell from dividing. Continued damage or stress causes mTOR production, which in turn activates protein synthesis and inactivates protein breakdown. Further stimulation of the cells leads to programmed cell death (apoptosis).
  • p16 is a protein involved in regulation of the cell cycle, by inhibiting the S phase (synthesis phase). It can be activated during ageing or in response to various stresses, such as DNA damage, oxidative stress or exposure to drugs. p16 is typically considered a tumor suppressor protein, causing a cell to become senescent in response to DNA damage and irreversibly preventing the cell from entering a hyperproliferative state. However, there has been some ambiguity in this regard, as some tumors show overexpression of p16, while other show downregulated expression. Evidence suggests that overexpression of p16 is some tumors results from a defective retinoblastoma protein (“Rb”).
  • Rb defective retinoblastoma protein
  • p16 acts on Rb to inhibit the S phase, and Rb downregulates p16, creating negative feedback.
  • Defective Rb fails to both inhibit the S phase and downregulate p16, thus resulting in overexpression of p16 in hyperproliferating cells (Romagosa, C. et al., p16 lnk4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors, Oncogene, Vol. 30, 2087-2097 (2011)).
  • Senescent cells are associated with secretion of many factors involved in intercellular signaling, including pro-inflammatory factors; secretion of these factors has been termed the senescence-associated secretory phenotype, or SASP (Freund, A. “Inflammatory networks during cellular senescence: causes and consequences” Trends Mol Med. 2010 May;16(5):238-46). Autoimmune diseases, such as Crohn’s disease and rheumatoid arthritis, are associated with chronic inflammation (Ferraccioli, G. et al.
  • Chronic inflammation may be characterized by the presence of pro- inflammatory factors at levels higher than baseline near the site of pathology, but lower than those found in acute inflammation.
  • these factors include TNF, IL-1a, IL-10, IL-5, IL-6, IL-8, IL-12, IL-23, CD2, CD3, CD20, CD22, CD52, CD80, CD86, C5 complement protein, BAFF, APRIL, IgE, a401 integrin and a4 7 integrin. Because senescent cells produce pro-inflammatory factors, removal of these cells alone produces a profound reduction in inflammation as well as the amount and concentration of pro-inflammatory factors.
  • ROS reactive oxygen species
  • the p16/Rb pathway leads to the induction of ROS, which in turn activates the protein kinase C delta creating a positive feedback loop that further enhance ROS, helping maintain the irreversible cell cycle arrest; it has even been suggested that exposing cancer cells to ROS might be effective to treat cancer by inducing cell phase arrest in hyperproliferating cells (Rayess, H. et al., Cellular senescence and tumor suppressor gene p16, Int J Cancer, Vol. 130, 1715-1725 (2012)).
  • mice that were treated to induce senescent cell elimination were found to have larger diameters of muscle fibers as compared to untreated mice. Treadmill exercise tests indicated that treatment also preserved muscle function. Continuous treatment of transgenic mice for removal of senescent cells had no negative side effects and selectively delayed age-related phenotypes that depend on cells. This data demonstrates that removal of senescent cells produces beneficial therapeutic effects and shows that these benefits may be achieved without adverse effects.
  • Vaccines have been widely used since their introduction by Edward Jenner in the 1770s to confer immunity against a wide range of diseases and afflictions.
  • Vaccine preparations contain a selected immunogenic agent capable of stimulating immunity to an antigen.
  • antigens are used as the immunogenic agent in vaccines, such as, for example, viruses, either killed or attenuated, and purified viral components.
  • Antigens used in the production of cancer vaccines include, for example, tumor-associated carbohydrate antigens (TACAs), dendritic cells, whole cells and viral vectors. Different techniques are employed to produce the desired amount and type of antigen being sought. For example, pathogenic viruses are grown either in eggs or cells. Recombinant DNA technology is often utilized to generate attenuated viruses for vaccines.
  • Vaccines may therefore be used to stimulate the production of antibodies in the body and provide immunity against antigens.
  • the immune system may destroy or remove cells that express the antigen.
  • the invention is a method of treating or preventing the onset of osteoarthritis comprising administering to a subject a composition comprising an anti-AGE antibody.
  • the invention is a method of treating or preventing the onset of osteoarthritis comprising administering to a subject a composition comprising a first anti-AGE antibody and a second anti-AGE antibody.
  • the second anti-AGE antibody is different from the first anti-AGE antibody.
  • the invention is a method of treating a subject with osteoarthritis comprising a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating osteoarthritis; followed by a second administering of the anti-AGE antibody.
  • the invention is use of an anti-AGE antibody for the manufacture of a medicament for treating or preventing the onset of osteoarthritis.
  • the invention is a composition comprising an anti-AGE antibody for use in treating or preventing the onset of osteoarthritis.
  • the invention is a composition for treating or preventing the onset of osteoarthritis comprising a first anti-AGE antibody, a second anti-AGE antibody and a pharmaceutically-acceptable carrier.
  • the first anti-AGE antibody is different from the second anti-AGE antibody.
  • the invention is a method of treating or preventing the onset of osteoarthritis comprising immunizing a subject in need thereof against AGE- modified proteins or peptides of a cell.
  • the invention is a method of treating a subject with osteoarthritis comprising administering a first vaccine comprising a first AGE antigen and, optionally, administering a second vaccine comprising a second AGE antigen.
  • the second AGE antigen is different from the first AGE antigen.
  • the invention is use of an AGE antigen for the manufacture of a medicament for treating or preventing the onset of osteoarthritis.
  • the invention is a composition comprising an AGE antigen for use in treating or preventing the onset of osteoarthritis.
  • peptide means a molecule composed of 2-50 amino acids.
  • protein means a molecule composed of more than 50 amino acids.
  • AGE-modified protein or peptide refers to modified proteins or peptides that are formed as the result of the reaction of sugars with protein side chains that further rearrange and form irreversible cross-links. This process begins with a reversible reaction between a reducing sugar and an amino group to form a Schiff base, which proceeds to form a covalently-bonded Amadori rearrangement product. Once formed, the Amadori product undergoes further rearrangement to produce AGEs.
  • AGE-modified proteins and antibodies to AGE-modified proteins are described in U.S. 5,702,704 to Bucala (“Bucala”) and U.S.
  • Al-Abed 6,380,165 to Al-Abed et al.
  • AGEs may be identified by the presence of AGE modifications (also referred to as AGE epitopes or AGE moieties) such as 2-(2-furoyl)-4(5)-(2-furanyl)-1H- imidazole (“FFI”); 5-hydroxymethyl-1-alkylpyrrole-2-carbaldehyde (“Pyrraline”); 1- alkyl-2-formyl-3,4-diglycosyl pyrrole (“AFGP”), a non-fluorescent model AGE; carboxymethyllysine; carboxyethyllysine; and pentosidine.
  • ALI another AGE, is described in Al-Abed.
  • AGE antigen means a substance that elicits an immune response against an AGE-modified protein or peptide of a cell.
  • the immune response against an AGE-modified protein or peptide of a cell does not include the production of antibodies to the non-AGE-modified protein or peptide.
  • an antibody that binds to an AGE-modified protein on a cell means an antibody, antibody fragment or other protein or peptide that binds to an AGE-modified protein or peptide which preferably includes a constant region of an antibody, where the protein or peptide which has been AGE-modified is a protein or peptide normally found bound on the surface of a cell, preferably a mammalian cell, more preferably a human, cat, dog, horse, camelid (for example, camel or alpaca), cattle, sheep, or goat cell.
  • an antibody that binds to an AGE-modified protein on a cell does not include an antibody or other protein which binds with the same specificity and selectivity to both the AGE-modified protein or peptide, and the same non-AGE- modified protein or peptide (that is, the presence of the AGE modification does not increase binding).
  • AGE-modified albumin is not an AGE-modified protein on a cell, because albumin is not a protein normally found bound on the surface of cells.
  • “An antibody that binds to an AGE-modified protein on a cell”, “anti-AGE antibody” or “AGE antibody” only includes those antibodies which lead to removal, destruction, or death of the cell.
  • antibodies which are conjugated, for example to a toxin, drug, or other chemical or particle Preferably, the antibodies are monoclonal antibodies, but polyclonal antibodies are also possible.
  • senescent cell means a cell which is in a state of proliferative arrest and expresses one or more biomarkers of senescence, such as activation of p16 lnk4a or expression of senescence-associated -galactosidase. Also included are cells which express one or more biomarkers of senescence, do not proliferate in vivo, but may proliferate in vitro under certain conditions, such as some satellite cells found in the muscles of ALS patients.
  • variant means a nucleotide, protein or amino acid sequence different from the specifically identified sequences, wherein one or more nucleotides, proteins or amino acid residues is deleted, substituted or added. Variants may be naturally-occurring allelic variants, or non-naturally-occurring variants. Variants of the identified sequences may retain some or all of the functional characteristics of the identified sequences.
  • percent (%) sequence identity is defined as the percentage of amino acid residues in a candidate sequence that are identical to the amino acid residues in a reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Preferably, % sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program is publicly available from Genentech, Inc.
  • ALIGN-2 (South San Francisco, GA), or may be compiled from the source code, which has been filed with user documentation in the U.S. Copyright Office and is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • the % sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows: 100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B.
  • FIG. 1 is a graph of the response versus time in an antibody binding experiment.
  • Osteoarthritis is typically considered a progressive disease caused by mechanical stress on joints over time and a natural result of aging, but its exact etiology is unknown.
  • Current research implicates cellular senescence in the onset and development of osteoarthritis.
  • a significant increase in the senescent biomarker p-jgink a has been found in osteoarthritic chondrocytes as compared to fetal chondrocytes or non-osteoarthritic chondrocytes of the same age (Zhang, H.W. et al., “Recovery of function in osteoarthritic chondrocytes induced by p16 lnk4a -specific siRNA in vitro”, Rheumatology, Vol.
  • Vaccination against AGE-modified proteins or peptides of a cell may also be used to control the presence of AGE-modified cells in a subject.
  • the continuous and virtually ubiquitous surveillance exercised by the immune system in the body in response to a vaccination allows maintaining low levels of AGE-modified cells in the body.
  • Vaccination against AGE-modified proteins or peptides of a cell removes or kills senescent cells. The process of senescent cell removal or destruction allows vaccination against AGE-modified proteins or peptides of a cell to be used to treat osteoarthritis.
  • an antibody that binds to an AGE-modified protein on a cell (“anti-AGE antibody” or “AGE antibody”) is known in the art. Examples include those described in U.S. 5,702,704 (Bucala) and U.S. 6,380,165 (Al-Abed et al.). Examples include an antibody that binds to one or more AGE-modified proteins having an AGE modification such as FFI, pyrraline, AFGP, ALI, carboxymethyllysine, carboxyethyllysine and pentosidine, and mixtures of such antibodies. Preferably, the antibody binds carboxymethyllysine-modified or carboxyethyllysine-modified proteins.
  • the antibody is non-immunogenic to the animal in which it will be used, such as non-immunogenic to humans; companion animals including cats, dogs and horses; and commercially important animals, such camels (or alpaca), cattle (bovine), sheep, and goats. More preferably, the antibody has the same species constant region as antibodies of the animal to reduce the immune response against the antibody, such as being humanized (for humans), felinized (for cats), caninized (for dogs), equuinized (for horses), carnalized (for camels or alpaca), bovinized (for cattle), ovinized (for sheep), or caperized (for goats).
  • the antibody is identical to that of the animal in which it will be used (except for the variable region), such as a human antibody, a cat antibody, a dog antibody, a horse antibody, a camel antibody, a bovine antibody, a sheep antibody or a goat antibody. Details of the constant regions and other parts of antibodies for these animals are described below.
  • the antibody may be monoclonal or polyclonal.
  • the antibody is a monoclonal antibody.
  • Particularly preferred anti-AGE antibodies include those which bind to proteins or peptides that exhibit a carboxymethyllysine or carboxyethyllysine AGE modification.
  • Carboxymethyllysine also known as N(epsilon)-(carboxymethyl)lysine, N(6)-carboxymethyllysine, or 2-Amino-6-(carboxymethylamino)hexanoic acid
  • carboxyethyllysine also known as N-epsilon-(carboxyethyl)lysine
  • CML- and CEL-modified proteins or peptides are recognized by the receptor RAGE which is expressed on a variety of cells. CML and CEL have been well-studied and CML- and CEL-related products are commercially available.
  • Cell Biolabs, Inc. sells CML-BSA antigens, CML polyclonal antibodies, CML immunoblot kits, and CML competitive ELISA kits (www.cellbiolabs.com/cml-assays) as well as CEL-BSA antigens and CEL competitive ELISA kits (www.cellbiolabs.com/cel-n- epsilon-carboxyethyl-lysine-assays-and-reagents).
  • a particularly preferred antibody includes the variable region of the commercially available mouse anti-glycation endproduct antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin, the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247), modified to have a human constant region (or the constant region of the animal into which it will be administered).
  • Commercially-available antibodies such as the carboxymethyl lysine antibody corresponding to catalog no. MAB3247 from R&D Systems, Inc., may be intended for diagnostic purposes and may contain material that is not suited for use in animals or humans.
  • commercially-available antibodies are purified and/or isolated prior to use in animals or humans to remove toxins or other potentially-harmful material.
  • the anti-AGE antibody has low rate of dissociation from the antibody-antigen complex, or kd (also referred to as kback or off-rate), preferably at most 9 x 10‘ 3 , 8 x
  • the anti-AGE antibody has a high affinity for the AGE-modified protein of a cell, which may be expressed as a low dissociation constant KD of at most 9 x 10’ 6 , 8 x 10’ 6 , 7 x 10’ 6 , 6 x 10’ 6 , 5 x 10’ 6 , 4 x 10- 6 or 3 x 10' 6 (M).
  • the binding properties of the anti-AGE antibody are similar to, the same as, or superior to the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247), illustrated in FIG. 1.
  • the anti-AGE antibody may destroy AGE-modified cells through antibodydependent cell-mediated cytotoxicity (ADCC).
  • ADCC is a mechanism of cell- mediated immune defense in which an effector cell of the immune system actively lyses a target cell whose membrane-surface antigens have been bound by specific antibodies.
  • ADCC may be mediated by natural killer (NK) cells, macrophages, neutrophils or eosinophils.
  • NK natural killer
  • the effector cells bind to the Fc portion of the bound antibody.
  • the anti-AGE antibody may also destroy AGE-modified cells through complement-dependent cytotoxicity (CDC). In CDC, the complement cascade of the immune system is triggered by an antibody binding to a target antigen.
  • CDC complement-dependent cytotoxicity
  • the anti-AGE antibody may be conjugated to an agent that causes the destruction of AGE-modified cells.
  • agents may be a toxin, a cytotoxic agent, magnetic nanoparticles, and magnetic spin-vortex discs.
  • a toxin such as pore-forming toxins (PFT) (Aroian R. etal., “Pore-Forming Toxins and Cellular Non-lmmune Defenses (CNIDs),” Current Opinion in Microbiology, 10:57-61 (2007)) conjugated to an anti-AGE antibody may be injected into a patient to selectively target and remove AGE-modified cells.
  • the anti-AGE antibody recognizes and binds to AGE-modified cells. Then, the toxin causes pore formation at the cell surface and subsequent cell removal through osmotic lysis.
  • Magnetic nanoparticles conjugated to the anti-AGE antibody may be injected into a patient to target and remove AGE-modified cells.
  • the magnetic nanoparticles can be heated by applying a magnetic field in order to selectively remove the AGE- modified cells.
  • Magnetic spin-vortex discs which are magnetized only when a magnetic field is applied to avoid self-aggregation that can block blood vessels, begin to spin when a magnetic field is applied, causing membrane disruption of target cells.
  • Magnetic spin-vortex discs, conjugated to anti-AGE antibodies specifically target AGE-modified cell types, without removing other cells.
  • Antibodies typically comprise two heavy chains and two light chains of polypeptides joined to form a "Y" shaped molecule. The constant region determines the mechanism used to target the antigen. The amino acid sequence in the tips of the "Y" (the variable region) varies among different antibodies. This variation gives the antibody its specificity for binding antigen.
  • variable region which includes the ends of the light and heavy chains, is further subdivided into hypervariable (HV - also sometimes referred to as complementarity determining regions, or CDRs) and framework (FR) regions.
  • HV hypervariable
  • FR framework
  • a humanized anti-AGE antibody according to the present invention may have the human constant region sequence of amino acids shown in SEQ ID NO: 22.
  • the heavy chain complementarity determining regions of the humanized anti-AGE antibody may have one or more of the protein sequences shown in SEQ ID NO: 23 (CDR1H), SEQ ID NO: 24 (CDR2H) and SEQ ID NO: 25 (CDR3H).
  • the light chain complementarity determining regions of the humanized anti-AGE antibody may have one or more of the protein sequences shown in SEQ ID NO: 26 (CDR1L), SEQ ID NO: 27 (CDR2L) and SEQ ID NO: 28 (CDR3L).
  • the heavy chain of human Homo sapiens) antibody immunoglobulin G1 may have or may include the protein sequence of SEQ ID NO: 1.
  • the variable domain of the heavy chain may have or may include the protein sequence of SEQ ID NO: 2.
  • the complementarity determining regions of the variable domain of the heavy chain (SEQ ID NO: 2) are shown in SEQ ID NO: 41, SEQ ID NO: 42 and SEQ ID NO: 43.
  • the kappa light chain of human (Homo sapiens) antibody immunoglobulin G1 may have or may include the protein sequence of SEQ ID NO: 3.
  • the variable domain of the kappa light chain may have or may include the protein sequence of SEQ ID NO: 4.
  • the arginine (Arg or R) residue at position 128 of SEQ ID NO: 4 may be omitted.
  • the complementarity determining regions of the variable domain of the light chain (SEQ ID NO: 4) are shown in SEQ ID NO: 44, SEQ ID NO: 45 and SEQ ID NO: 46.
  • the variable regions may be codon-optimized, synthesized and cloned into expression vectors containing human immunoglobulin G1 constant regions.
  • the variable regions may be used in the humanization of non-human antibodies.
  • the antibody heavy chain may be encoded by the DNA sequence of SEQ ID NO: 12, a murine anti-AGE immunoglobulin G2b heavy chain.
  • the protein sequence of the murine anti-AGE immunoglobulin G2b heavy chain encoded by SEQ ID NO: 12 is shown in SEQ ID NO: 16.
  • the variable region of the murine antibody is shown in SEQ ID NO: 20, which corresponds to positions 25-142 of SEQ ID NO: 16.
  • the antibody heavy chain may alternatively be encoded by the DNA sequence of SEQ ID NO: 13, a chimeric anti-AGE human immunoglobulin G1 heavy chain.
  • the protein sequence of the chimeric anti-AGE human immunoglobulin G1 heavy chain encoded by SEQ ID NO: 13 is shown in SEQ ID NO: 17.
  • the chimeric anti-AGE human immunoglobulin includes the murine variable region of SEQ ID NO: 20 in positions 25-142.
  • the antibody light chain may be encoded by the DNA sequence of SEQ ID NO: 14, a murine anti-AGE kappa light chain.
  • the protein sequence of the murine anti-AGE kappa light chain encoded by SEQ ID NO: 14 is shown in SEQ ID NO: 18.
  • the variable region of the murine antibody is shown in SEQ ID NO: 21, which corresponds to positions 21-132 of SEQ ID NO: 18.
  • the antibody light chain may alternatively be encoded by the DNA sequence of SEQ ID NO: 15, a chimeric anti- AGE human kappa light chain.
  • the protein sequence of the chimeric anti-AGE human kappa light chain encoded by SEQ ID NO: 15 is shown in SEQ ID NO: 19.
  • the chimeric anti-AGE human immunoglobulin includes the murine variable region of SEQ ID NO: 21 in positions 21-132.
  • a humanized anti-AGE antibody according to the present invention may have or may include one or more humanized heavy chains or humanized light chains.
  • a humanized heavy chain may be encoded by the DNA sequence of SEQ ID NO: 30, 32 or 34.
  • the protein sequences of the humanized heavy chains encoded by SEQ ID NOs: 30, 32 and 34 are shown in SEQ ID NOs: 29, 31 and 33, respectively.
  • a humanized light chain may be encoded by the DNA sequence of SEQ ID NO: 36, 38 or 40.
  • the protein sequences of the humanized light chains encoded by SEQ ID NOs: 36, 38 and 40 are shown in SEQ ID NOs: 35, 37 and 39, respectively.
  • the humanized anti-AGE antibody maximizes the amount of human sequence while retaining the original antibody specificity.
  • a complete humanized antibody may be constructed that contains a heavy chain having a protein sequence chosen from SEQ ID NOs: 29, 31 and 33 and a light chain having a protein sequence chosen from SEQ ID NOs: 35, 37 and 39.
  • the protein sequence of an antibody from a non-human species may be modified to include the variable domain of the heavy chain having the sequence shown in SEQ ID NO: 2 or the kappa light chain having the sequence shown in SEQ ID NO: 4.
  • the non-human species may be a companion animal, such as the domestic cat or domestic dog, or livestock, such as cattle, the horse or the camel. Preferably, the non-human species is not the mouse.
  • the heavy chain of the horse (Equus caballus) antibody immunoglobulin gamma 4 may have or may include the protein sequence of SEQ ID NO: 5 (EMBL/GenBank accession number AY445518).
  • the heavy chain of the horse (Equus caballus) antibody immunoglobulin delta may have or may include the protein sequence of SEQ ID NO: 6 (EMBL/GenBank accession number AY631942).
  • the heavy chain of the dog (Canis familiaris) antibody immunoglobulin A may have or may include the protein sequence of SEQ ID NO: 7 (GenBank accession number L36871).
  • the heavy chain of the dog (Canis familiaris) antibody immunoglobulin E may have or may include the protein sequence of SEQ ID NO: 8 (GenBank accession number L36872).
  • the heavy chain of the cat (Felis catus) antibody immunoglobulin G2 may have or may include the protein sequence of SEQ ID NO: 9 (DDBJ/EMBL/GenBank accession number KF811175).
  • Animals of the camelid family such as camels (Camelus dromedarius and Camelus bactrianus), llamas (Lama glama, Lama pacos and Lama vicugna), alpacas (Vicugna pacos) and guanacos (Lama guanicoe), have a unique antibody that is not found in other mammals.
  • camelids also have heavy chain immunoglobulin G antibodies that do not contain light chains and exist as heavy chain dimers.
  • variable domain of a camelid heavy chain antibody is known as the VHH.
  • the camelid heavy chain antibodies lack the heavy chain CH1 domain and have a hinge region that is not found in other species.
  • the variable region of the Arabian camel (Camelus dromedarius) single-domain antibody may have or may include the protein sequence of SEQ ID NO: 10 (GenBank accession number AJ245148).
  • the variable region of the heavy chain of the Arabian camel (Camelus dromedarius) tetrameric immunoglobulin may have or may include the protein sequence of SEQ ID NO: 11 (GenBank accession number AJ245184).
  • IgNAR immunoglobulin new antigen receptor
  • VNAR variable domain of an IgNAR
  • the protein sequences of additional non-human species may be readily found in online databases, such as the International ImMunoGeneTics Information System (www.imgt.org), the European Bioinformatics Institute (www.ebi.ac.uk), the DNA Databank of Japan (ddbj.nig.ac.jp/arsa) or the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov).
  • online databases such as the International ImMunoGeneTics Information System (www.imgt.org), the European Bioinformatics Institute (www.ebi.ac.uk), the DNA Databank of Japan (ddbj.nig.ac.jp/arsa) or the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov).
  • An anti-AGE antibody or a variant thereof may include a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 20, including post-translational modifications thereof.
  • a variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity may contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-AGE antibody including that sequence retains the ability to bind to AGE.
  • substitutions, insertions, or deletions may occur in regions outside the variable region.
  • An anti-AGE antibody or a variant thereof may include a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 21 , including post-translational modifications thereof.
  • a variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity may contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-AGE antibody including that sequence retains the ability to bind to AGE.
  • substitutions, insertions, or deletions may occur in regions outside the variable region.
  • the antibody may have the complementarity determining regions of commercially available mouse anti-glycation end-product antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin (CML-KLH), the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247).
  • CML-KLH keyhole limpet hemocyanin
  • CDN carboxymethyl lysine MAb
  • the antibody may have or may include constant regions which permit destruction of targeted cells by a subject’s immune system.
  • Bi-specific antibodies which are anti-AGE antibodies directed to two different epitopes, may also be used. Such antibodies will have a variable region (or complementary determining region) from those of one anti-AGE antibody, and a variable region (or complementary determining region) from a different antibody.
  • Antibody fragments may be used in place of whole antibodies.
  • immunoglobulin G may be broken down into smaller fragments by digestion with enzymes. Papain digestion cleaves the N-terminal side of inter-heavy chain disulfide bridges to produce Fab fragments.
  • Fab fragments include the light chain and one of the two N-terminal domains of the heavy chain (also known as the Fd fragment).
  • Pepsin digestion cleaves the C-terminal side of the inter-heavy chain disulfide bridges to produce F(ab’)2 fragments.
  • F(ab’)2 fragments include both light chains and the two N-terminal domains linked by disulfide bridges.
  • Pepsin digestion may also form the Fv (fragment variable) and Fc (fragment crystallizable) fragments.
  • the Fv fragment contains the two N-terminal variable domains.
  • the Fc fragment contains the domains which interact with immunoglobulin receptors on cells and with the initial elements of the complement cascade.
  • Pepsin may also cleave immunoglobulin G before the third constant domain of the heavy chain (CH3) to produce a large fragment F(abc) and a small fragment pFc’.
  • Antibody fragments may alternatively be produced recombinantly. Preferably, such antibody fragments are conjugated to an agent that causes the destruction of AGE-modified cells.
  • antibodies can be produced using well-known methods.
  • polyclonal antibodies pAbs
  • pAbs polyclonal antibodies
  • an immunogen and if desired, an adjuvant.
  • the immunogen (and adjuvant) is injected in a mammal by a subcutaneous or intraperitoneal injection.
  • the immunogen may be an AGE-modified protein of a cell, such as AGE-antithrombin III, AGE-calmodulin, AGE-insulin, AGE- ceruloplasmin, AGE-collagen, AGE-cathepsin B, AGE-albumin such as AGE-bovine serum albumin (AGE-BSA), AGE-human serum albumin and ovalbumin, AGE- crystallin, AGE-plasminogen activator, AGE-endothelial plasma membrane protein, AGE-aldehyde reductase, AGE-transferrin, AGE-fibrin, AGE-copper/zinc SOD, AGE- apo B, AGE-fibronectin, AGE-pancreatic ribose, AGE-apo A-l and II, AGE- hemoglobin, AGE-Na + /K + -ATPase, AGE-plasminogen, AGE-myelin, AGE-lysozyme,
  • AGE-modified cells such as AGE-modified erythrocytes, whole, lysed, or partially digested, may also be used as AGE antigens.
  • adjuvants include Freund’s complete, monophosphoryl Lipid A synthetic-trehalose dicorynomycolate, aluminum hydroxide (alum), heat shock proteins HSP 70 or HSP96, squalene emulsion containing monophosphoryl lipid A, a2-macroglobulin and surface active substances, including oil emulsions, pleuronic polyols, polyanions and dinitrophenol.
  • an immunogen may be conjugated to a polypeptide that is immunogenic in the host, such as keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, cholera toxin, labile enterotoxin, silica particles or soybean trypsin inhibitor.
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • bovine thyroglobulin bovine thyroglobulin
  • cholera toxin cholera toxin
  • labile enterotoxin silica particles
  • silica particles silica particles
  • soybean trypsin inhibitor e.g., soybean trypsin inhibitor.
  • Monoclonal antibodies may also be made by immunizing a host or lymphocytes from a host, harvesting the mAb-secreting (or potentially secreting) lymphocytes, fusing those lymphocytes to immortalized cells (for example, myeloma cells), and selecting those cells that secrete the desired mAb.
  • Other techniques may be used, such as the EBV-hybridoma technique.
  • chimeric antibodies that are substantially human (humanized) or substantially “ized” to another animal (such as cat, dog, horse, camel or alpaca, cattle, sheep, or goat) at the amino acid level.
  • the mAbs may be purified from the culture medium or ascites fluid by conventional procedures, such as protein A-sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, ammonium sulfate precipitation or affinity chromatography.
  • human monoclonal antibodies can be generated by immunization of transgenic mice containing a third copy IgG human trans-loci and silenced endogenous mouse Ig loci or using human-transgenic mice. Production of humanized monoclonal antibodies and fragments thereof can also be generated through phage display technologies.
  • a "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • Preferred examples of such carriers or diluents include water, saline, Ringer’s solutions and dextrose solution. Supplementary active compounds can also be incorporated into the compositions.
  • Solutions and suspensions used for parenteral administration can include a sterile diluent, such as water for injection, saline solution, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • the antibodies may be administered by injection, such as by intravenous injection or intra-articular injection into an affected joint.
  • Pharmaceutical compositions suitable for injection include sterile aqueous solutions or dispersions for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • Suitable carriers include physiological saline, bacteriostatic water, CREMOPHOR EL® (BASF; Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid so as to be administered using a syringe.
  • compositions should be stable during manufacture and storage and must be preserved against contamination from microorganisms such as bacteria and fungi.
  • Various antibacterial and anti-fungal agents for example, parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal, can contain microorganism contamination.
  • Isotonic agents such as sugars, polyalcohols, such as manitol, sorbitol, and sodium chloride can be included in the composition.
  • Compositions that can delay absorption include agents such as aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating antibodies, and optionally other therapeutic components, in the required amount in an appropriate solvent with one or a combination of ingredients as required, followed by sterilization. Methods of preparation of sterile solids for the preparation of sterile injectable solutions include vacuum drying and freeze-drying to yield a solid.
  • the antibodies may be delivered as an aerosol spray from a nebulizer or a pressurized container that contains a suitable propellant, for example, a gas such as carbon dioxide.
  • a suitable propellant for example, a gas such as carbon dioxide.
  • Antibodies may also be delivered via inhalation as a dry powder, for example using the iSPERSETM inhaled drug delivery platform (PULMATRIX, Lexington, Mass.).
  • the use of anti-AGE antibodies which are chicken antibodies (IgY) may be non-immunogenic in a variety of animals, including humans, when administered by inhalation.
  • An appropriate dosage level of each type of antibody will generally be about
  • the dosage level will be about 0.1 to about 250 mg/kg; more preferably about 0.5 to about 100 mg/kg.
  • a suitable dosage level may be about 0.01 to 250 mg/kg, about 0.05 to 100 mg/kg, or about 0.1 to 50 mg/kg. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg.
  • each type of antibody may be administered on a regimen of 1 to 4 times per day, such as once or twice per day, antibodies typically have a long half-life in vivo. Accordingly, each type of antibody may be administered once a day, once a week, once every two or three weeks, once a month, or once every 60 to 90 days.
  • a subject that receives administration of an anti-AGE antibody may be tested to determine if the administration has been effective to treat osteoarthritis by monitoring symptoms of osteoarthritis overtime, such as pain level, stiffness and joint functionality. These factors may be measured directly or may be analyzed according to a standard medical grading scale, such as the WOMAC scale. Osteoarthritis may also be evaluated radiographically, such as by analysis of X-rays or magnetic resonance imaging (MRI) radiographs. Diagnostic imaging can reveal signs of osteoarthritis including joint space narrowing, increased subchondral bone density, increased subchondral stenosis (increased bone formation), subchondral cyst formation, osteophyte formation and hard tissue enlargement.
  • MRI magnetic resonance imaging
  • Unit dosage forms can be created to facilitate administration and dosage uniformity.
  • Unit dosage form refers to physically discrete units suited as single dosages for the subject to be treated, containing a therapeutically effective quantity of one or more types of antibodies in association with the required pharmaceutical carrier.
  • the unit dosage form is in a sealed container and is sterile.
  • Vaccines against AGE-modified proteins or peptides contain an AGE antigen, an adjuvant, optional preservatives and optional excipients.
  • AGE antigens include AGE-modified proteins or peptides such as AGE-antithrombin III, AGE-calmodulin, AGE-insulin, AGE-ceruloplasmin, AGE-collagen, AGE-cathepsin B, AGE-albumin such as AGE-bovine serum albumin (AGE-BSA), AGE-human serum albumin and ovalbumin, AGE-crystallin, AGE-plasminogen activator, AGE- endothelial plasma membrane protein, AGE-aldehyde reductase, AGE-transferrin, AGE-fibrin, AGE-copper/zinc SOD, AGE-apo B, AGE-fibronectin, AGE-pancreatic ribose, AGE-apo A-l and II, AGE-hemo
  • AGE- modified cells such as AGE-modified erythrocytes, whole, lysed, or partially digested, may also be used as AGE antigens.
  • Suitable AGE antigens also include proteins or peptides that exhibit AGE modifications (also referred to as AGE epitopes or AGE moieties) such as carboxymethyllysine (CML), carboxyethyllysine (CEL), pentosidine, pyrraline, FFI, AFGP and ALL
  • the AGE antigen may be an AGE- protein conjugate, such as AGE conjugated to keyhole limpet hemocyanin (AGE- KLH). Further details of some of these AGE-modified proteins or peptides and their preparation are described in Bucala.
  • Particularly preferred AGE antigens include proteins or peptides that exhibit a carboxymethyllysine or carboxyethyllysine AGE modification.
  • Carboxymethyllysine also known as N(epsilon)-(carboxymethyl)lysine, N(6)-carboxymethyllysine, or 2- Amino-6-(carboxymethylamino)hexanoic acid
  • carboxyethyllysine also known as N-epsilon-(carboxyethyl)lysine
  • proteins or peptides and lipids as a result of oxidative stress and chemical glycation and have been correlated with juvenile genetic disorders.
  • CML- and CEL-modified proteins or peptides are recognized by the receptor RAGE which is expressed on a variety of cells.
  • CML and CEL have been well-studied and CML- and CEL-related products are commercially available.
  • Cell Biolabs, Inc. sells CM L-BSA antigens, CML polyclonal antibodies, CML immunoblot kits, and CML competitive ELISA kits (www.cellbiolabs.com/cml-assays) as well as CEL-BSA antigens and CEL competitive ELISA kits (www.cellbiolabs.com/cel-n-epsilon-carboxyethyl-lysine- assays-and-reagents).
  • AGE antigens may be conjugated to carrier proteins to enhance antibody production in a subject. Antigens that are not sufficiently immunogenic alone may require a suitable carrier protein to stimulate a response from the immune system.
  • suitable carrier proteins include keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, cholera toxin, labile enterotoxin, silica particles and soybean trypsin inhibitor.
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • bovine thyroglobulin cholera toxin
  • labile enterotoxin silica particles
  • soybean trypsin inhibitor e.g., the carrier protein is KLH (AGE-KLH).
  • KLH has been extensively studied and has been identified as an effective carrier protein in experimental cancer vaccines.
  • Preferred AGE antigen-carrier protein conjugates include CML-KLH and CEL-KLH.
  • Immunity is a long-term immune response, either cellular or humoral.
  • a cellular immune response is activated when an antigen is presented, preferably with a co-stimulator to a T-cell which causes it to differentiate and produce cytokines.
  • the cells involved in the generation of the cellular immune response are two classes of T-helper (Th) cells, Th1 and Th2.
  • Th1 cells stimulate B cells to produce predominantly antibodies of the lgG2A isotype, which activates the complement cascade and binds the Fc receptors of macrophages, while Th2 cells stimulate B cells to produce lgG1 isotype antibodies in mice, lgG4 isotype antibodies in humans, and IgE isotype antibodies.
  • the human body also contains “professional” antigen-presenting cells such as dendritic cells, macrophages, and B cells.
  • a humoral immune response is triggered when a B cell selectively binds to an antigen and begins to proliferate, leading to the production of a clonal population of cells that produce antibodies that specifically recognize that antigen and which may differentiate into antibody-secreting cells, referred to as plasma-cells or memory-B cells.
  • Antibodies are molecules produced by B-cells that bind a specific antigen.
  • the antigen-antibody complex triggers several responses, either cell-mediated, for example by natural killers (NK) or macrophages, or serum-mediated, for example by activating the complement system, a complex of several serum proteins that act sequentially in a cascade that result in the lysis of the target cell.
  • Immunological adjuvants are the component(s) of a vaccine which augment the immune response to the immunogenic agent Adjuvants function by attracting macrophages to the immunogenic agent and then presenting the agent to the regional lymph nodes to initiate an effective antigenic response. Adjuvants may also act as carriers themselves for the immunogenic agent. Adjuvants may induce an inflammatory response, which may play an important role in initiating the immune response.
  • Adjuvants include mineral compounds such as aluminum salts, oil emulsions, bacterial products, liposomes, immunostimulating complexes and squalene.
  • Aluminum compounds are the most widely used adjuvants in human and veterinary vaccines. These aluminum compounds include aluminum salts such as aluminum phosphate (AIPO4) and aluminum hydroxide (AI(OH)3) compounds, typically in the form of gels, and are generically referred to in the field of vaccine immunological adjuvants as "alum.”
  • Aluminum hydroxide is a poorly crystalline aluminum oxyhydroxide having the structure of the mineral boehmite.
  • Aluminum phosphate is an amorphous aluminum hydroxy phosphate.
  • Negatively charged species for example, negatively charged antigens
  • positively charged species for example, positively charged antigens
  • Aluminum compounds provide a depot of antigen at the site of administration, thereby providing a gradual and continuous release of antigen to stimulate antibody production. Aluminum compounds tend to more effectively stimulate a cellular response mediated by Th2, rather than Th1 cells.
  • Emulsion adjuvants include water-in-oil emulsions (for example, Freund's adjuvants, such as killed mycobacteria in oil emulsion) and oil-in-water emulsions (for example, MF-59).
  • Emulsion adjuvants include an immunogenic component, for example squalene (MF-59) or mannide oleate (Incomplete Freund's Adjuvants), which can induce an elevated humoral response, increased T cell proliferation, cytotoxic lymphocytes and cell-mediated immunity.
  • Liposomal or vesicular adjuvants include paucilamellar lipid vesicles
  • Paucilamellar vesicles can be prepared by mixing, under high pressure or shear conditions, a lipid phase comprising a nonphospholipid material (for example, an amphiphile surfactant; see U.S. Pat Nos.
  • a sterol optionally a sterol, and any water-immiscible oily material to be encapsulated in the vesicles (for example, an oil such as squalene oil and an oil-soluble or oil-suspended antigen); and an aqueous phase such as water, saline, buffer or any other aqueous solution used to hydrate the lipids.
  • a sterol optionally a sterol, and any water-immiscible oily material to be encapsulated in the vesicles
  • an oil such as squalene oil and an oil-soluble or oil-suspended antigen
  • an aqueous phase such as water, saline, buffer or any other aqueous solution used to hydrate the lipids.
  • Liposomal or vesicular adjuvants are believed to promote contact of the antigen with immune cells, for example by fusion of the vesicle to the immune cell membrane, and preferentially stimulate
  • adjuvants include Mycobacterium bovis bacillus Calmette- Guerin (BCG), quill-saponin and unmethylated CpG dinucleotides (CpG motifs). Additional adjuvants are described in U.S. Patent Application Publication Pub. No. US 2010/0226932 (September 9, 2010) and Jiang, Z-H. et al. “Synthetic vaccines: the role of adjuvants in immune targeting”, Current Medicinal Chemistry, Vol. 10(15), pp. 1423-39 (2003). Preferable adjuvants include Freund’s complete adjuvant and Freund’s incomplete adjuvant.
  • the vaccine may optionally include one or more preservatives, such as antioxidants, antibacterial and antimicrobial agents, as well as combinations thereof.
  • preservatives such as antioxidants, antibacterial and antimicrobial agents, as well as combinations thereof.
  • examples include benzethonium chloride, ethylenediamine-tetraacetic acid sodium (EDTA), thimerosal, phenol, 2-phenoxyethanol, formaldehyde and formalin; antibacterial agents such as amphotericin B, chlortetracycline, gentamicin, neomycin, polymyxin B and streptomycin; antimicrobial surfactants such as polyoxyethylene-9, 10-nonyl phenol (Triton N-101 , octoxynol-9), sodium deoxycholate and polyoxyethylated octyl phenol (Triton X-I00).
  • the production and packaging of the vaccine may eliminate the need for a preservative. For example, a vaccine that has been sterilize
  • compositions include pharmaceutically acceptable excipients, such as stabilizers, thickening agents, toxin detoxifiers, diluents, pH adjusters, tonicity adjustors, surfactants, antifoaming agents, protein stabilizers, dyes and solvents.
  • pharmaceutically acceptable excipients such as stabilizers, thickening agents, toxin detoxifiers, diluents, pH adjusters, tonicity adjustors, surfactants, antifoaming agents, protein stabilizers, dyes and solvents.
  • excipients examples include hydrochloric acid, phosphate buffers, sodium acetate, sodium bicarbonate, sodium borate, sodium citrate, sodium hydroxide, potassium chloride, potassium chloride, sodium chloride, polydimethylsilozone, brilliant green, phenol red (phenolsulfon-phthalein), glycine, glycerin, sorbitol, histidine, monosodium glutamate, potassium glutamate, sucrose, urea, lactose, gelatin, sorbitol, polysorbate 20, polysorbate 80 and glutaraldehyde.
  • hydrochloric acid phosphate buffers, sodium acetate, sodium bicarbonate, sodium borate, sodium citrate, sodium hydroxide, potassium chloride, potassium chloride, sodium chloride, polydimethylsilozone, brilliant green, phenol red (phenolsulfon-phthalein), glycine, glycerin, sorbitol, histidine, monosodium glutamate, potassium glut
  • the vaccine may contain from 1 pg to 100 mg of at least one AGE antigen, including 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 400, 800 or 1000 pg, or 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80 or 90 mg.
  • the amount used for a single injection corresponds to a unit dosage.
  • the vaccine may be provided in unit dosage form or in multidosage form, such as 2-100 or 2-10 doses.
  • the unit dosages may be provided in a vial with a septum, or in a syringe with or without a needle.
  • the vaccine may be administered intravenously, subdermally or intraperitoneally.
  • the vaccine is sterile.
  • the vaccine may be administered one or more times, such as 1 to 10 times, including 2, 3, 4, 5, 6, 7, 8 or 9 times, and may be administered over a period of time ranging from 1 week to 1 year, 2-10 weeks or 2-10 months. Furthermore, booster vaccinations may be desirable, over the course of 1 year to 20 years, including 2, 5, 10 and 15 years.
  • a subject that receives a vaccine for AGE-modified proteins or peptides of a cell may be tested to determine if he or she has developed an immunity to the AGE- modified proteins or peptides. Suitable tests may include blood tests for detecting the presence of an antibody, such as immunoassays or antibody titers. Alternatively, an immunity to AGE-modified proteins or peptides may be determined by monitoring osteoarthritis symptoms of a subject over time. For example, a baseline knee stiffness of a subject may be measured followed by administration of the vaccine for AGE-modified proteins or peptides of a cell.
  • Immunity to AGE-modified proteins or peptides may be determined by periodically measuring the knee stiffness of the subject and comparing the subsequent measurements to the baseline measurement.
  • a subject may be considered to have developed an immunity to AGE-modified proteins or peptides if he or she demonstrates an improvement in knee stiffness between subsequent measurements or over time.
  • An immunity to AGE-modified proteins or peptides may also be determined by monitoring the concentration and/or number of senescent cells over time.
  • a subject may also be tested to determine if the vaccination has been effective to treat osteoarthritis.
  • the effectiveness of the vaccination may be determined by vaccinating a subject followed by periodically obtaining X-rays and/or MRI images of the subject over time or measuring the concentration and/or number of senescent cells. Vaccination and subsequent testing may be repeated until the desired therapeutic result is achieved.
  • the vaccination process may be designed to provide immunity against multiple AGE moieties. A single AGE antigen may induce the production of AGE antibodies which are capable of binding to multiple AGE moieties. Alternatively, the vaccine may contain multiple AGE antigens. In addition, a subject may receive multiple vaccines, where each vaccine contains a different AGE antigen.
  • Any mammal that could develop osteoarthritis may be treated by the methods herein described. Humans are a preferred mammal for treatment. Other mammals that may be treated include mice, rats, goats, sheep, cows, horses and companion animals, such as dogs or cats. Alternatively, any of the mammals or subjects identified above may be excluded from the patient population in need of treatment for osteoarthritis.
  • a subject may be identified as having osteoarthritis or in need of treatment if he or she exhibits any of the classical osteoarthritis symptoms, such as joint pain, stiffness, reduced range of motion, joint swelling or weakness or numbness of the arms and legs.
  • a subject may also be identified as in need of treatment based on analysis of diagnostic images such as X-rays and MRI radiographs for the presence of joint space narrowing, increased subchondral bone density, increased subchondral stenosis (increased bone formation), subchondral cyst formation, osteophyte formation or hard tissue enlargement.
  • subjects may be identified as in need of treatment due to specific physical changes that are known risk factors for osteoarthritis such as a direct injury to a joint or having legs of unequal length.
  • Subjects may also be identified as in need of treatment based on the presence of a pathological condition associated with AGEs such as, for example, atherosclerosis, inflammation, retinopathy, nephropathy, stroke, endothelial cell dysfunction, neurodegenerative disorders or cancer.
  • Positions 16-133 of the above amino acid sequence correspond to SEQ ID NO: 2. Positions 46-50 of the above amino acid sequence correspond to SEQ ID NO: 41. Positions 65-81 of the above amino acid sequence correspond to SEQ ID NO: 42. Positions 114-122 of the above amino acid sequence correspond to SEQ ID NO: 43.
  • Positions 16-128 of the above amino acid sequence correspond to SEQ ID NO: 4.
  • the arginine (Arg or R) residue at position 128 of SEQ ID NO: 4 may be omitted.
  • Positions 39-54 of the above amino acid sequence correspond to SEQ ID NO: 44.
  • Positions 70-76 of the above amino acid sequence correspond to
  • the alanine residue at position 123 of the above amino acid sequence may optionally be replaced with a serine residue.
  • the tyrosine residue at position 124 of the above amino acid sequence may optionally be replaced with a phenylalanine residue.
  • Positions 25-142 of the above amino acid sequence correspond to SEQ ID NO: 20.
  • SEQ ID NO: 20 may optionally include the substitutions at positions 123 and 124.
  • SEQ ID NO: 20 may optionally contain one additional lysine residue after the terminal valine residue.
  • the one-letter amino acid sequence that corresponds to SEQ ID NO: 17 is shown below:
  • Positions 21-132 of the above amino acid sequence correspond to SEQ ID NO: 21.
  • KCCVECPPCP APPVAGPSVF LFPPKPKDTL MISRTPEVTC VWDVSHEDP 160 170 180 190
  • VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK
  • SEQ ID NO: 23 The one-letter amino acid sequence that corresponds to SEQ ID NO: 23 is SYTMGVS.
  • the one-letter amino acid sequence that corresponds to SEQ ID NO: 24 is TISSGGGSTYYPDSVKG.
  • the one-letter amino acid sequence that corresponds to SEQ ID NO: 25 is QGGWLPPFAX, where X may be any naturally occurring amino acid.
  • LVSNLES [113] The one-leter amino acid sequence that corresponds to SEQ ID NO: 28 is QHIRELTRS.
  • the one-leter amino acid sequence that corresponds to SEQ ID NO: 29 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHWVRQAPGQGLEWMGEISPTNGRAYYNQKFQGRVTMTVDKSTNTVYMELSS LRSEDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTA ALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQ TYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMIS RTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVL HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLT CLVK
  • the one-letter amino acid sequence that corresponds to SEQ ID NO: 31 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHVWRQAPGQGLEWMGEISPTNGRAYYNAKFQGRVTMTVDKSTNTAYMELSS LRSEDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTA ALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQ TYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMIS RTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVL HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLT CLVKGFYPS
  • the DNA sequence that corresponds to SEQ ID NO: 32 is ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTG AAGAAACCTGGCGCCTCCGTGAGGTGTCCTGCAAGGCTTCCGGCTACCTGTTC ACCACCTACTGGATGCACTGGGTGCGACAGGCCCCTGGACAGGGCCTGGAAT GGATGGGCGAGATCTCCCCTACCAACGGCAGAGCCTACTACAACCAAAATTCC AGGGCAGAGTGACCATGACCGTGGACAAGTCCACCAACACCGCTTACATGGAA CTGTCCTCCCTGCGGAGCGAGGACACCGCCGTGTACTACTGCGCTAGAGCCTA CGGCAACTACGATTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCCTC TGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGC ACCAG
  • the one-leter amino acid sequence that corresponds to SEQ ID NO: 33 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHWVRQAPGQGLEWMGEISPTNGRAYYNAKFQGRVTMTVDKSINTAYMELSRL RSDDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAA LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQT YICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMISR TPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVLH QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLTC LVKGFY
  • the DNA sequence that corresponds to SEQ ID NO: 34 is ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTG AAGAAACCTGGCGCCTCCGTGAGGTGTCCTGCAAGGCTTCCGGCTACCTGTTC ACCACCTACTGGATGCACTGGGTGCGACAGGCCCCTGGACAGGGCCTGGAAT GGATGGGCGAGATCTCCCCTACCAACGGCAGAGCCTACTACAACCAAAATTCC AGGGCAGAGTGACCATGACCGTGGACAAGTCCATCAACACCGCTTACATGGAA CTGTCCAGACTGCGGAGCGATGACACCGCCGTGTACTACTGCTAGAGCCTA CGGCAACTACGATTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCCTC TGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCAGCAGCAAGAGC ACCAGCG
  • the one-leter amino acid sequence that corresponds to SEQ ID NO: 35 is METDTLLLWVLLLWVPGSTGDWMTQSPLSLPVTLGQPASISCRSSQSLVNSNGNT FLQWYQQRPGQSPRLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY CSQSTHVPPTFGGGTVEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAK VQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG LSSPVTKSFNRGEC.
  • the DNA sequence that corresponds to SEQ ID NO: 36 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCCTGCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCTCCCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCAGCAGCGGCCTGGCCAGAGCCCCAGAC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCG GCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CGGAGGCACCAAGTGGAAATCAAGCGGACCGTGGCCGCCCAGCGTGTTCA TCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGCTGG
  • the one-letter amino acid sequence that corresponds to SEQ ID NO: 37 is METDTLLLWVLLLWVPGSTGDWMTQSPLSLPVTLGQPASISCRSRQSLVNSNGN TFLQWYQQRPGQSPRLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVY YCSQSTHVPPTFGGGTVEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREA KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGEC.
  • the DNA sequence that corresponds to SEQ ID NO: 38 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCCTGCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCAGGCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCAGCAGCGGCCTGGCCAGAGCCCCAGAC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCG GCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CGGAGGCACCAAGTGGAAATCAAGCGGACCGTGGCCGCCCAGCGTGTTCA TCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGCTGG
  • the one-leter amino acid sequence that corresponds to SEQ ID NO: 39 is METDTLLLWVLLLWVPGSTGDWMTQSPLSSPVTLGQPASISCRSSQSLVNSNGN TFLQWYHQRPGQPPRLLIYKVSLRFSGVPDRFSGSGAGKDFTLKISRVEAEDVGVY YCSQSTHVPPTFGQGTLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREA KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGEC.
  • the DNA sequence that corresponds to SEQ ID NO: 40 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCAGTCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCTCCCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCACCAGCGGCCTGGCCAGCCTCCCAGACT GCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCGG CTCTGGCGCTGGCAAGGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CCAGGGCACCAACTGGAAATCAAGCGGACCGTGGCCGCCCAGCGTGTTCA TCTTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGTTCA TCTTCCCAG
  • Example 1 In vivo study of the administration of anti-glycation end-product antibody
  • the antibody was administered to the aged CD1(ICR) mouse (Charles River Laboratories), twice daily by intravenous injection, once a week, for three weeks (Days 1 , 8 and 15), followed by a 10 week treatment-free period.
  • the test antibody was a commercially available mouse anti-glycation end-product antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin, the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247).
  • a control reference of physiological saline was used in the control animals.
  • mice referred to as “young” were 8 weeks old, while mice referred to as “old” were 88 weeks ( ⁇ 2 days) old. No adverse events were noted from the administration of the antibody.
  • the different groups of animals used in the study are shown in Table 1.
  • Pre Subset of animals euthanized prior to treatment start for collection of adipose tissue.
  • Example 2 Affinity and kinetics of test antibody
  • Example 1 The affinity and kinetics of the test antibody used in Example 1 were analyzed using Na,Na-bis(carboxymethyl)-L-lysine trifluoroacetate salt (Sigma-Aldrich, St. Louis, MO) as a model substrate for an AGE-modified protein of a cell. Label-free interaction analysis was carried out on a BIACORETM T200 (GE Healthcare, Pittsburgh, PA), using a Series S sensor chip CM5 (GE Healthcare, Pittsburgh, PA), with Fc1 set as blank, and Fc2 immodilized with the test antibody (molecular weigh of 150,000 Da).
  • the running buffer was a HBS-EP buffer (10 mM HEPES, 150 mM NaCI, 3 mM EDTA and 0.05% P-20, pH of 7.4), at a temperature of 25 °C.
  • Software was BIACORETM T200 evaluation software, version 2.0. A double reference (Fc2-1 and only buffer injection), was used in the analysis, and the data was fitted to a Langmuir 1:1 binding model.
  • Example 3 Construction and production of murine anti-AGE lgG2b antibody and chimeric anti-AGE lgG1 antibody
  • Murine and chimeric human anti-AGE antibodies were prepared.
  • the DNA sequence of murine anti-AGE antibody lgG2b heavy chain is shown in SEQ ID NO: 12.
  • the DNA sequence of chimeric human anti-AGE antibody lgG1 heavy chain is shown in SEQ ID NO: 13.
  • the DNA sequence of murine anti-AGE antibody kappa light chain is shown in SEQ ID NO: 14.
  • the DNA sequence of chimeric human anti- AGE antibody kappa light chain is shown in SEQ ID NO: 15.
  • the gene sequences were synthesized and cloned into high expression mammalian vectors. The sequences were codon optimized. Completed constructs were sequence confirmed before proceeding to transfection.
  • HEK293 cells were seeded in a shake flask one day before transfection, and were grown using serum-free chemically defined media.
  • the DNA expression constructs were transiently transfected into 0.03 liters of suspension HEK293 cells. After 20 hours, cells were sampled to obtain the viabilities and viable cell counts, and titers were measured (Octet QKe, ForteBio). Additional readings were taken throughout the transient transfection production runs. The cultures were harvested on day 5, and an additional sample for each was measured for cell density, viability and titer.
  • Example 3 The binding of the murine (parental) and chimeric anti-AGE antibodies described in Example 3 was investigated by a direct binding ELISA.
  • An anti- carboxymethyl lysine (CML) antibody (R&D Systems, MAB3247) was used as a control.
  • CML was conjugated to KLH (CML-KLH) and both CML and CML-KLH were coated overnight onto an ELISA plate.
  • HRP-goat anti-mouse Fc was used to detect the control and murine (parental) anti-AGE antibodies.
  • HRP-goat anti-human Fc was used to detect the chimeric anti-AGE antibody.
  • the antigens were diluted to 1 pg/mL in 1x phosphate buffer at pH 6.5.
  • a 96- well microtiter ELISA plate was coated with 100 pL/well of the diluted antigen and let sit at 4°C overnight. The plate was blocked with 1x PBS, 2.5% BSA and allowed to sit for 1-2 hours the next morning at room temperature.
  • the antibody samples were prepared in serial dilutions with 1x PBS, 1% BSA with the starting concentration of 50 pg/mL. Secondary antibodies were diluted 1 :5,000. 100 pL of the antibody dilutions was applied to each well. The plate was incubated at room temperature for 0.5-1 hour on a microplate shaker.
  • the plate was washed 3 times with 1x PBS. 100 pL/well diluted HRP-conjugated goat anti-human Fc secondary antibody was applied to the wells. The plate was incubated for 1 hour on a microplate shaker. The plate was then washed 3 times with 1x PBS. 100 pL HRP substrate TMB was added to each well to develop the plate. After 3-5 minutes elapsed, the reaction was terminated by adding 100 pL of 1 N HCL A second direct binding ELISA was performed with only CML coating. The absorbance at OD450 was read using a microplate reader.
  • the OD450 absorbance raw data for the CML-only ELISA is shown in the plate map below. 24 of the 96 wells in the well plate were used. Blank wells in the plate map indicate unused wells.
  • Control AGE AGE [156] The control and chimeric anti-AGE antibodies showed binding to both CML and CML-KLH.
  • the murine (parental) anti-AGE antibody showed very weak to no binding to either CML or CML-KLH.
  • Data from repeated ELISA confirms binding of the control and chimeric anti-AGE to CML. All buffer control showed negative signal.
  • Humanized antibodies were designed by creating multiple hybrid sequences that fuse select parts of the parental (mouse) antibody sequence with the human framework sequences. Acceptor frameworks were identified based on the overall sequence identity across the framework, matching interface position, similarly classed CDR canonical positions, and presence of N-glycosylation sites that would have to be removed. Three humanized light chains and three humanized heavy chains were designed based on two different heavy and light chain human acceptor frameworks. The amino acid sequences of the heavy chains are shown in SEQ ID NO: 29, 31 and 33, which are encoded by the DNA sequences shown in SEQ ID NO: 30, 32 and 34, respectively.
  • the amino acid sequences of the light chains are shown in SEQ ID NO: 35, 37 and 39, which are encoded by the DNA sequences shown in SEQ ID NO: 36, 38 and 40, respectively.
  • the humanized sequences were methodically analyzed by eye and computer modeling to isolate the sequences that would most likely retain antigen binding. The goal was to maximize the amount of human sequence in the final humanized antibodies while retaining the original antibody specificity.
  • the light and heavy humanized chains could be combined to create nine variant fully humanized antibodies.
  • the three heavy chains and three light chains were analyzed to determine their humanness.
  • Antibody humanness scores were calculated according to the method described in Gao, S. H., et al., “Monoclonal antibody humanness score and its applications”, BMC Biotechnology, 13:55 (July 5, 2013).
  • the humanness score represents how human-like an antibody variable region sequence looks. For heavy chains a score of 79 or above is indicative of looking human-like; for light chains a score of 86 or above is indicative of looking human-like.
  • the humanness of the three heavy chains, three light chains, a parental (mouse) heavy chain and a parental (mouse) light chain are shown below in Table 6:
  • variable region sequences were designed by first synthesizing the variable region sequences. The sequences were optimized for expression in mammalian cells. These variable region sequences were then cloned into expression vectors that already contain human Fc domains; for the heavy chain, the lgG1 was used.
  • the binding of the humanized antibodies may be evaluated, for example, by dose-dependent binding ELISA or cell-based binding assay.
  • Example 6 An AGE-RNAse containing vaccine in a human subject.
  • AGE-RNAse is prepared by incubating RNAse in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-RNAse solution is dialyzed and the protein content is measured.
  • Aluminum hydroxide or aluminum phosphate, as an adjuvant, is added to 100 pg of the AGE-RNAse.
  • Formaldehyde or formalin is added as a preservative to the preparation. Ascorbic acid is added as an antioxidant.
  • the vaccine also includes phosphate buffer to adjust the pH and glycine as a protein stabilizer. The composition is injected intravenously into a subject with osteoarthritis.
  • Example 7 Injection regimen for an AGE-RNAse containing vaccine in a human subject
  • Example 6 The same vaccine as described in Example 6 is injected intra-articularly into a subject with osteoarthritis.
  • the titer of antibodies to AGE-RNAse is determined by ELISA after two weeks. Additional injections are performed after three weeks and six weeks, respectively. Further titer determination is performed two weeks after each injection.
  • Example 8 An AGE-hemoglobin containing vaccine in a human subject.
  • AGE-hemoglobin is prepared by incubating human hemoglobin in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-hemoglobin solution is dialyzed and the protein content is measured. All vaccine components are the same as in Example 6, except AGE-hemoglobin is substituted for AGE-RNAse. Administration is carried out as in Example 6, or as in Example 7.
  • Example 9 An AGE-human serum albumin containing vaccine in a human subject.
  • AGE-human serum albumin is prepared by incubating human serum albumin in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-human serum albumin solution is dialyzed and the protein content is measured. All vaccine components are the same as in Example 6, except AGE-human serum albumin is substituted for AGE-RNAse. Administration is carried out as in Example 6, or as in Example 7.
  • Example 10 Carboxymethyllysine-modified protein vaccine for a human subject (prophetic)
  • a vaccine is prepared by combining a carboxymethyllysine-modified protein as an AGE antigen, aluminum hydroxide as an adjuvant, formaldehyde as a preservative, ascorbic acid as an antioxidant, a phosphate buffer to adjust the pH of the vaccine and glycine as a protein stabilizer.
  • the vaccine is injected subcutaneously into a subject with osteoarthritis.
  • Example 11 Carboxyethyllysine-modified peptide vaccine for a human subject (prophetic)
  • a vaccine is prepared by combining a carboxyethyllysine-modified peptide conjugated to KLH as an AGE antigen, aluminum hydroxide as an adjuvant, formaldehyde as a preservative, ascorbic acid as an antioxidant, a phosphate buffer to adjust the pH of the vaccine and glycine as a protein stabilizer.
  • the vaccine is injected subcutaneously into a subject with osteoarthritis.
  • Example 12 In vivo study of the administration of a carboxymethyl lysine monoclonal antibody
  • 4T1 murine breast tumor cells (ATCC CRL-2539) were cultured in RPMI 1640 medium containing 10% fetal bovine serum, 2 mM glutamine, 25 pg/mL gentamicin, 100 units/mL penicillin G Na and 100 pg/mL streptomycin sulfate. Tumor cells were maintained in tissue culture flasks in a humidified incubator at 37 °C in an atmosphere of 5% CO2 and 95% air. [181] The cultured breast cancer cells were then implanted in the mice.
  • PBS phosphate buffered saline
  • the four treatment groups are shown in Table 8 below:
  • An anti-carboxymethyl lysine monoclonal antibody was used as a therapeutic agent 250 mg of carboxymethyl lysine monoclonal antibody was obtained from R&D Systems (Minneapolis, MN). Dosing solutions of the carboxymethyl lysine monoclonal antibody were prepared at 1 and 0.5 mg/mL in a vehicle (PBS) to provide the active dosages of 10 and 5 pg/g, respectively, in a dosing volume of 10 mL/kg. Dosing solutions were stored at 4 °C protected from light.
  • PBS vehicle
  • %TGI (1- MTVtreated/MTVcontrol) X 100.
  • the ability of the anti-carboxymethyl lysine antibody to inhibit cachexia was determined by comparing the weights of the lungs and gastrocnemius muscles for Groups 1-3. Tissue weights were also normalized to 100 g body weight.
  • Treatment efficacy was also evaluated by the incidence and magnitude of regression responses observed during the study.
  • Treatment may cause partial regression (PR) or complete regression (CR) of the tumor in an animal.
  • PR partial regression
  • CR complete regression
  • the tumor volume was 50% or less of its Day 1 volume for three consecutive measurements during the course of the study, and equal to or greater than 13.5 mm 3 for one or more of these three measurements.
  • the tumor volume was less than 13.5 mm 3 for three consecutive measurements during the course of the study.
  • mice In vivo studies are carried out in mice to study the effect of treatment with anti- AGE antibodies and vaccination with AGE-KLH on osteoarthritis.
  • Male C57/BL6 mice are 8-10 weeks old on Day 1 of the study.
  • the mice are separated into five treatment groups: (1) control; (2) vehicle only administered intravenously; (3) anti- AGE antibody at 10 pg/g dose administered intravenously; (4) anti-AGE antibody at 10 pg/g dose administered intra-articularly; and (5) 10 pg AGE-KLH administered as a vaccine intraperitoneally.
  • Osteoarthritis is induced in Groups 2-5 by medial meniscal destabilization of the right hind paw knee.
  • Group 1 is a non-arthritic control is sham operated on by exposure of the left hind paw medial meniscotibial ligament with no transection. All animals are anesthetized with isoflurane, shaved and prepared for surgery.
  • destabilization is achieved by the complete rupture of the medial meniscus ligament on the left joint.
  • the knee capsule is closed with absorbable suture material. Skin is closed with a subcutaneous suture of absorbable suture and surgical glue.
  • Buprenorphine (0.3mg/ml) is provided at a dose level of 0.1mg/kg presurgery and 8-12 hours post first injection.
  • Dosing begins one week after the surgery.
  • the dosing volume is 0.200 mL per 20 grams of body weight (10 mL/kg), and is scaled to the body weight of each individual animal.
  • Group 2 receives phosphate-buffered saline (PBS) delivered intravenously.
  • Group 3 receives 10pg/g of an anti-AGE antibody twice daily for 21 days delivered intravenously.
  • Group 4 receives 10pg/g of an anti- AGE antibody twice daily for 21 days delivered intra-articularly into the knee that was operated on.
  • Group 5 receives 10pg of AGE-KLH in Freunds complete adjuvant intraperitoneally one week prior to inducing osteoarthritis, followed by a 10pg/g booster injection of the vaccine four weeks after surgery.
  • DMB dynamic weight bearing
  • the animals in Groups 1 and 5 are sacrificed at week 16.
  • the blood is collected for an antibody titer assay, such as the THERMOFISHER® EASY- TITER® Mouse IgG Assay, to determine the titer of antibody in the mice specific for anti-AGE antibodies.
  • An equal number of animals in Groups 2-4 are sacrificed at weeks 4, 8 and 16.
  • Half of the mice in each sacrificed group are analyzed for histology and half are analyzed for p16INK4a qRT PCR.
  • p16INK4a is measured in articular cartilage (chondrocytes) of the animals sacrificed.
  • the p16INK4a qRT PCR is preserved for qRT PCR analysis.
  • Osteoarthritis severity is also measured by evaluating samples of the knee joints. Sample of the right and left whole knee joints from all mice are collected and fixed in 10% NBF, then decalcified and embedded in paraffin wax. Three non- consecutive coronal sections are taken for the right knee joint and another three non- consecutive coronal sections are taken for the left knee joint for each staining, providing 6 slides per animal for each stain for a total of 12 slides per animal. The sections are scored for disease severity (cartilage/bone with osteophytes and synovial membrane) by a board certified veterinary pathologist using a semi- quantitative grading system. Scores are reported with statistical analysis.
  • the anti-AGE antibody will specifically bind to senescent chondrocytes and allow the immune system to destroy those cells. Similarly, vaccination with an AGE- KLH antigen will allow the murine immune system to target and remove senescent chondrocytes. Killing and removing senescent chondrocytes will prevent the development of osteoarthritis.
  • Example 14 Anti-AGE antibodies bind to senescent chondrocytes in vitro
  • Senescent chondrocytes were obtained from osteoarthritic joints.
  • Anti-AGE antibodies bound to the senescent chondrocytes in vitro. These results suggest that anti-AGE antibodies could be administered therapeutically in vivo to treat osteoarthritis by specifically targeting senescent chondrocytes.
  • Example 15 Summary of Jeon et al., 2017, which is incorporated herein by reference
  • senescent cells were selectively removed. Selective killing of senescent cells was achieved by administering ganciclovir to the p16-3MR mice, which is effective due to the fact that the p16-3MR transgene includes a truncated form of herpes simplex virus 1 thymidine kinase (HSV-TK). The clearance resulted in a reduction in p16 INK4a expression. Clearance of senescent cells inhibited articular cartilage erosion.
  • HSV-TK herpes simplex virus 1 thymidine kinase

Abstract

A method of treating osteoarthritis comprises administering to a subject a composition comprising an anti-AGE antibody. A composition for treating osteoarthritis comprises a first anti-AGE antibody, a second anti-AGE antibody and a pharmaceutically acceptable carrier. The first anti-AGE antibody is different from the second anti-AGE antibody. The anti-AGE antibody may be administered intra-articularly into an affected joint. A method of treating or preventing the onset of osteoarthritis comprises immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.

Description

METHODS AND COMPOSITIONS FOR TREATING OSTEOARTHRITIS USING ANTI-AGE ANTIBODIES OR AGE ANTIGENS
BACKGROUND
[01] Arthritis is the general term for the more than 100 different diseases and disorders that affect joints or articulations of the body. Osteoarthritis is the most common type of arthritis and affects approximately 30 million adults in the United States (U.S. Centers for Disease Control and Prevention). Worldwide, it is estimated that 10% of all individuals over age 60 have significant clinical problems that can be attributed to osteoarthritis (World Health Organization).
[02] Osteoarthritis results from the deterioration of joint cartilage, which is composed of specialized cells known as chondrocytes. Chondrocytes produce a collagenous extracellular matrix containing collagen and proteoglycans. The loss or breakdown of cartilage increases friction between the bones of the joint, which can lead to the growth of bone spurs (osteophytes) or the release of cartilage and bone fragments into the joint space. Symptoms of osteoarthritis include joint pain, stiffness, reduced range of motion, joint swelling and weakness or numbness of the arms and legs. Osteoarthritis may develop in any joint, but is most commonly seen in the hands and fingers, knees, hips, feet, neck and back.
[03] Current treatments for osteoarthritis focus on reducing risk factors that contribute to osteoarthritis and managing symptoms of osteoarthritis. Lifestyle changes that can reduce risk factors include exercise and weight control. Osteoarthritis-associated pain is often treated with over-the-counter analgesics, such as acetaminophen and nonsteroidal anti-inflammatory drugs (NSAIDs), or prescription pain medicines, such as opiates and COX-2 inhibitors. Local treatment of an affected joint may involve intra-articular injection of corticosteroids, hyaluronic acid or hyaluronic acid substitutes. In more severe cases, surgery may be required to repair or replace the affected joint. These therapies fail to address the underlying cause of osteoarthritis, the deterioration of cartilage. [04] Senescent cells are cells that are partially-functional or non-functional and are in a state of proliferative arrest. Senescence is a distinct state of a cell, and is associated with biomarkers, such as activation of the biomarker p16lnk4a, and expression of p-galactosidase. Senescence begins with damage or stress (such as overstimulation by growth factors) of cells.
[05] Advanced glycation end-products (AGEs; also referred to as AGE-modified proteins, or glycation end-products) arise from a non-enzymatic reaction of sugars with protein side-chains (Ando, K. et al., Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation, Biochem Biophys Res Commun., Vol. 258, 123, 125 (1999)). This process begins with a reversible reaction between the reducing sugar and the amino group to form a Schiff base, which proceeds to form a covalently-bonded Amadori rearrangement product. Once formed, the Amadori product undergoes further rearrangement to produce AGEs. Hyperglycemia, caused by diabetes mellitus (DM), and oxidative stress promote this post-translational modification of membrane proteins (Lindsey JB, et al., “Receptor For Advanced Glycation End-Products (RAGE) and soluble RAGE (sRAGE): Cardiovascular Implications,” Diabetes Vascular Disease Research, Vol. 6(1), 7-14, (2009)). AGEs may also be formed from other processes. For example, the advanced glycation end product, Ne-(carboxymethyl)lysine, is a product of both lipid peroxidation and glycoxidation reactions. AGEs have been associated with several pathological conditions including diabetic complications, inflammation, retinopathy, nephropathy, atherosclerosis, stroke, endothelial cell dysfunction, and neurodegenerative disorders (Bierhaus A, “AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept,” Cardiovasc Res, Vol. 37(3), 586-600 (1998)).
[06] AGE-modified proteins are also a marker of senescent cells. This association between glycation end-product and senescence is well known in the art. See, for example, Gruber, L. (WO 2009/143411, 26 Nov. 2009), Ando, K. et al. (Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation, Biochem Biophys Res Common., Vol. 258, 123, 125 (1999)), Ahmed, E.K. etal. (“Protein Modification and Replicative Senescence of Wl- 38 Human Embryonic Fibroblasts” Aging Cells, vol. 9, 252, 260 (2010)), Vlassara, H. et al. (Advanced Glycosylation Endproducts on Erythrocyte Cell Surface Induce Receptor-Mediated Phagocytosis by Macrophages, J. Exp. Med., Vol. 166, 539, 545 (1987)) and Vlassara et al. (“High-affinity-receptor-mediated Uptake and Degradation of Glucose-modified Proteins: A Potential Mechanism for the Removal of Senescent Macromolecules” Proc. Natl. Acad. Sci. USAI, Vol. 82, 5588, 5591 (1985)). Furthermore, Ahmed, E.K. et al. indicates that glycation end-products are “one of the major causes of spontaneous damage to cellular and extracellular proteins" (Ahmed, E.K. et al., see above, page 353). Accordingly, the accumulation of glycation endproducts is associated with senescence and lack of function.
[07] The damage or stress that causes cellular senescence also negatively impacts mitochondrial DNA in the cells to cause them to produce free radicals which react with sugars in the cell to form methyl glyoxal (MG). MG in turn reacts with proteins or lipids to generate advanced glycation end products. In the case of the protein component lysine, MG reacts to form carboxymethyllysine, which is an AGE.
[08] Damage or stress to mitochondrial DNA also sets off a DNA damage response which induces the cell to produce cell cycle blocking proteins. These blocking proteins prevent the cell from dividing. Continued damage or stress causes mTOR production, which in turn activates protein synthesis and inactivates protein breakdown. Further stimulation of the cells leads to programmed cell death (apoptosis).
[09] p16 is a protein involved in regulation of the cell cycle, by inhibiting the S phase (synthesis phase). It can be activated during ageing or in response to various stresses, such as DNA damage, oxidative stress or exposure to drugs. p16 is typically considered a tumor suppressor protein, causing a cell to become senescent in response to DNA damage and irreversibly preventing the cell from entering a hyperproliferative state. However, there has been some ambiguity in this regard, as some tumors show overexpression of p16, while other show downregulated expression. Evidence suggests that overexpression of p16 is some tumors results from a defective retinoblastoma protein (“Rb”). p16 acts on Rb to inhibit the S phase, and Rb downregulates p16, creating negative feedback. Defective Rb fails to both inhibit the S phase and downregulate p16, thus resulting in overexpression of p16 in hyperproliferating cells (Romagosa, C. et al., p16lnk4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors, Oncogene, Vol. 30, 2087-2097 (2011)).
[10] Senescent cells are associated with secretion of many factors involved in intercellular signaling, including pro-inflammatory factors; secretion of these factors has been termed the senescence-associated secretory phenotype, or SASP (Freund, A. “Inflammatory networks during cellular senescence: causes and consequences” Trends Mol Med. 2010 May;16(5):238-46). Autoimmune diseases, such as Crohn’s disease and rheumatoid arthritis, are associated with chronic inflammation (Ferraccioli, G. et al. “Interleukin-1 and lnterleukin-6 in Arthritis Animal Models: Roles in the Early Phase of Transition from Acute to Chronic Inflammation and Relevance for Human Rheumatoid Arthritis” Mol Med. 2010 Nov-Dec; 16(11-12): 552-557). Chronic inflammation may be characterized by the presence of pro- inflammatory factors at levels higher than baseline near the site of pathology, but lower than those found in acute inflammation. Examples of these factors include TNF, IL-1a, IL-10, IL-5, IL-6, IL-8, IL-12, IL-23, CD2, CD3, CD20, CD22, CD52, CD80, CD86, C5 complement protein, BAFF, APRIL, IgE, a401 integrin and a4 7 integrin. Because senescent cells produce pro-inflammatory factors, removal of these cells alone produces a profound reduction in inflammation as well as the amount and concentration of pro-inflammatory factors.
[11] Senescent cells secrete reactive oxygen species (“ROS”) as part of the SASP. ROS is believed to play an important role in maintaining senescence of cells. The secretion of ROS creates a bystander effect, where senescent cells induce senescence in neighboring cells: ROS create the very cellular damage known to activate p16 expression, leading to senescence (Nelson, G., A senescent cell bystander effect: senescence-induced senescence, Aging Cell, Vo. 11 , 345-349 (2012)). The p16/Rb pathway leads to the induction of ROS, which in turn activates the protein kinase C delta creating a positive feedback loop that further enhance ROS, helping maintain the irreversible cell cycle arrest; it has even been suggested that exposing cancer cells to ROS might be effective to treat cancer by inducing cell phase arrest in hyperproliferating cells (Rayess, H. et al., Cellular senescence and tumor suppressor gene p16, Int J Cancer, Vol. 130, 1715-1725 (2012)).
[12] Recent research demonstrates the therapeutic benefits of removing senescent cells. In vivo animal studies at the Mayo Clinic in Rochester, Minnesota, found that elimination of senescent cells in transgenic mice carrying a biomarker for elimination delayed age-related disorders associated with cellular senescence. Eliminating senescent cells in fat and muscle tissues substantially delayed the onset of sarcopenia and cataracts and reduced senescence indicators in skeletal muscle and the eye (Baker, D. J. et al., “Clearance of p16lnk4a-positive senescent cells delays ageing-associated disorders”, Nature, Vol. 479, pp. 232-236, (2011)). Mice that were treated to induce senescent cell elimination were found to have larger diameters of muscle fibers as compared to untreated mice. Treadmill exercise tests indicated that treatment also preserved muscle function. Continuous treatment of transgenic mice for removal of senescent cells had no negative side effects and selectively delayed age-related phenotypes that depend on cells. This data demonstrates that removal of senescent cells produces beneficial therapeutic effects and shows that these benefits may be achieved without adverse effects.
[13] Additional In vivo animal studies in mice found that senescent cells using senolytic agents treats aging-related disorders and atherosclerosis. Short-term treatment with senolytic drugs in chronologically aged or progeroid mice alleviated several aging-related phenotypes (Zhu, Y. et al., “The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs”, Aging Cell, vol. 14, pp. 644-658 (2015)). Long-term treatment with senolytic drugs improved vasomotor function in mice with established atherosclerosis and reduced intimal plaque calcification (Roos, C.M. et al., “Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice”, Aging Cell (2016)). This data further demonstrates the benefits of removing senescent cells.
[14] Vaccines have been widely used since their introduction by Edward Jenner in the 1770s to confer immunity against a wide range of diseases and afflictions. Vaccine preparations contain a selected immunogenic agent capable of stimulating immunity to an antigen. Typically, antigens are used as the immunogenic agent in vaccines, such as, for example, viruses, either killed or attenuated, and purified viral components. Antigens used in the production of cancer vaccines include, for example, tumor-associated carbohydrate antigens (TACAs), dendritic cells, whole cells and viral vectors. Different techniques are employed to produce the desired amount and type of antigen being sought. For example, pathogenic viruses are grown either in eggs or cells. Recombinant DNA technology is often utilized to generate attenuated viruses for vaccines.
[15] Vaccines may therefore be used to stimulate the production of antibodies in the body and provide immunity against antigens. When an antigen is introduced to a subject that has been vaccinated and developed immunity to that antigen, the immune system may destroy or remove cells that express the antigen.
SUMMARY
[16] In a first aspect, the invention is a method of treating or preventing the onset of osteoarthritis comprising administering to a subject a composition comprising an anti-AGE antibody.
[17] In a second aspect, the invention is a method of treating or preventing the onset of osteoarthritis comprising administering to a subject a composition comprising a first anti-AGE antibody and a second anti-AGE antibody. The second anti-AGE antibody is different from the first anti-AGE antibody.
[18] In a third aspect, the invention is a method of treating a subject with osteoarthritis comprising a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating osteoarthritis; followed by a second administering of the anti-AGE antibody.
[19] In a fourth aspect, the invention is use of an anti-AGE antibody for the manufacture of a medicament for treating or preventing the onset of osteoarthritis. [20] In a fifth aspect, the invention is a composition comprising an anti-AGE antibody for use in treating or preventing the onset of osteoarthritis.
[21] In a sixth aspect, the invention is a composition for treating or preventing the onset of osteoarthritis comprising a first anti-AGE antibody, a second anti-AGE antibody and a pharmaceutically-acceptable carrier. The first anti-AGE antibody is different from the second anti-AGE antibody.
[22] In a seventh aspect, the invention is a method of treating or preventing the onset of osteoarthritis comprising immunizing a subject in need thereof against AGE- modified proteins or peptides of a cell.
[23] In an eighth aspect, the invention is a method of treating a subject with osteoarthritis comprising administering a first vaccine comprising a first AGE antigen and, optionally, administering a second vaccine comprising a second AGE antigen. The second AGE antigen is different from the first AGE antigen.
[24] In a ninth aspect, the invention is use of an AGE antigen for the manufacture of a medicament for treating or preventing the onset of osteoarthritis.
[25] In a tenth aspect, the invention is a composition comprising an AGE antigen for use in treating or preventing the onset of osteoarthritis.
[26] DEFINITIONS
[27] The term “peptide” means a molecule composed of 2-50 amino acids.
[28] The term "protein" means a molecule composed of more than 50 amino acids.
[29] The terms “advanced glycation end-product”, “AGE”, “AGE-modified protein or peptide" and “glycation end-product” refer to modified proteins or peptides that are formed as the result of the reaction of sugars with protein side chains that further rearrange and form irreversible cross-links. This process begins with a reversible reaction between a reducing sugar and an amino group to form a Schiff base, which proceeds to form a covalently-bonded Amadori rearrangement product. Once formed, the Amadori product undergoes further rearrangement to produce AGEs. AGE-modified proteins and antibodies to AGE-modified proteins are described in U.S. 5,702,704 to Bucala (“Bucala”) and U.S. 6,380,165 to Al-Abed et al. (“Al-Abed”). Glycated proteins or peptides that have not undergone the necessary rearrangement to form AGEs, such as N-deoxyfructosyllysine found on glycated albumin, are not AGEs. AGEs may be identified by the presence of AGE modifications (also referred to as AGE epitopes or AGE moieties) such as 2-(2-furoyl)-4(5)-(2-furanyl)-1H- imidazole ("FFI"); 5-hydroxymethyl-1-alkylpyrrole-2-carbaldehyde ("Pyrraline"); 1- alkyl-2-formyl-3,4-diglycosyl pyrrole ("AFGP"), a non-fluorescent model AGE; carboxymethyllysine; carboxyethyllysine; and pentosidine. ALI, another AGE, is described in Al-Abed.
[30] The term “AGE antigen” means a substance that elicits an immune response against an AGE-modified protein or peptide of a cell. The immune response against an AGE-modified protein or peptide of a cell does not include the production of antibodies to the non-AGE-modified protein or peptide.
[31] “An antibody that binds to an AGE-modified protein on a cell”, “anti-AGE antibody” or “AGE antibody” means an antibody, antibody fragment or other protein or peptide that binds to an AGE-modified protein or peptide which preferably includes a constant region of an antibody, where the protein or peptide which has been AGE-modified is a protein or peptide normally found bound on the surface of a cell, preferably a mammalian cell, more preferably a human, cat, dog, horse, camelid (for example, camel or alpaca), cattle, sheep, or goat cell. “An antibody that binds to an AGE-modified protein on a cell", “anti-AGE antibody” or “AGE antibody” does not include an antibody or other protein which binds with the same specificity and selectivity to both the AGE-modified protein or peptide, and the same non-AGE- modified protein or peptide (that is, the presence of the AGE modification does not increase binding). AGE-modified albumin is not an AGE-modified protein on a cell, because albumin is not a protein normally found bound on the surface of cells. “An antibody that binds to an AGE-modified protein on a cell”, “anti-AGE antibody” or “AGE antibody” only includes those antibodies which lead to removal, destruction, or death of the cell. Also included are antibodies which are conjugated, for example to a toxin, drug, or other chemical or particle. Preferably, the antibodies are monoclonal antibodies, but polyclonal antibodies are also possible.
[321 The term “senescent cell” means a cell which is in a state of proliferative arrest and expresses one or more biomarkers of senescence, such as activation of p16lnk4a or expression of senescence-associated -galactosidase. Also included are cells which express one or more biomarkers of senescence, do not proliferate in vivo, but may proliferate in vitro under certain conditions, such as some satellite cells found in the muscles of ALS patients.
[33] The term “variant’ means a nucleotide, protein or amino acid sequence different from the specifically identified sequences, wherein one or more nucleotides, proteins or amino acid residues is deleted, substituted or added. Variants may be naturally-occurring allelic variants, or non-naturally-occurring variants. Variants of the identified sequences may retain some or all of the functional characteristics of the identified sequences.
[34] The term "percent (%) sequence identity" is defined as the percentage of amino acid residues in a candidate sequence that are identical to the amino acid residues in a reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Preferably, % sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program is publicly available from Genentech, Inc. (South San Francisco, GA), or may be compiled from the source code, which has been filed with user documentation in the U.S. Copyright Office and is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. [35] In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: 100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. Where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained using the ALIGN-2 computer program.
BRIEF DESCRIPTION OF THE DRAWING
[36] FIG. 1 is a graph of the response versus time in an antibody binding experiment.
DETAILED DESCRIPTION
[37] Osteoarthritis is typically considered a progressive disease caused by mechanical stress on joints over time and a natural result of aging, but its exact etiology is unknown. Current research implicates cellular senescence in the onset and development of osteoarthritis. A significant increase in the senescent biomarker p-jgink a has been found in osteoarthritic chondrocytes as compared to fetal chondrocytes or non-osteoarthritic chondrocytes of the same age (Zhang, H.W. et al., “Recovery of function in osteoarthritic chondrocytes induced by p16lnk4a-specific siRNA in vitro”, Rheumatology, Vol. 43, No. 5, pp. 555-568 (2004)). Multiple studies strongly suggest that progressive chondrocyte senescence contributes to cartilage degeneration by decreasing the ability of chondrocytes to maintain and repair tissue (Martin, J.A. et al., “Aging, articular cartilage chondrocyte senescence and osteoarthritis”, Biogerontology, Vol. 3, pp. 254-264 (2002)). In vivo and in vitro studies indicate that erosion of chondrocyte telomere length and mitochondrial degeneration due to oxidative damage promote chondrocyte senescence (Martin et al.). A recent in vivo study at the Mayo Clinic found that injection of senescent cells into the knees of mice can induce osteoarthritis, while injection of non-senescent cells does not (Xu, M. et al., “Transplanted senescent cells induce an osteoarthritislike condition in mice”, Journals of Gerontology: Biological Sciences, Vol. 00, No. 00, pp. 1-6 (August 10, 2016)).
[38] Inflammation and tissue degradation are hallmarks of osteoarthritis, that can be mediated by senescence-associated secretory phenotype (SASP) (Jeon, O.H. et al., “Senescent cells and osteoarthritis: a painful connection”, Journal of Clinical Investigation, Vol. 128, No. 4, pp. 1229-1237 (2018)). Studies have found new evidence of the causative role of senescent cells in the development of post- traumatic osteoarthritis (Jeon et al., 2018). Accordingly, there is a causal link between cellular senescence and osteoarthritis.
[39] In vitro studies have shown that reducing senescent features by silencing p16lnk4a with small interfering ribonucleic acid (siRNA) can promote recovery of function in osteoarthritic chondrocytes (Zhang et al.). The therapeutic benefits of removing senescent cells has been demonstrated in atherosclerosis and in age- related diseases, such as sarcopenia. The identification of a link between cellular senescence and osteoarthritis allows for similar treatment possibilities. The present invention uses enhanced clearance of cells expressing AGE-modified proteins or peptides (AGE-modified cells) to treat, ameliorate or prevent the onset of osteoarthritis. This may be accomplished by administering anti-AGE antibodies to a subject.
[40] The selective elimination of senescent cells accumulated in the articular cartilage and synovium attenuated the development of post-traumatic osteoarthritis, reduced pain and increased cartilage development (Jeon, O.H. et al., “Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment’, Nature Medicine, Vol. 23, pp. 775-781 (2017)). Intra-articular injection of a senolytic molecule that selectively killed senescent cells validated these results in transgenic, non-transgenic and aged mice. (Jeon, O.H. et al., 2017).
[41] Vaccination against AGE-modified proteins or peptides of a cell may also be used to control the presence of AGE-modified cells in a subject. The continuous and virtually ubiquitous surveillance exercised by the immune system in the body in response to a vaccination allows maintaining low levels of AGE-modified cells in the body. Vaccination against AGE-modified proteins or peptides of a cell removes or kills senescent cells. The process of senescent cell removal or destruction allows vaccination against AGE-modified proteins or peptides of a cell to be used to treat osteoarthritis.
[42] An antibody that binds to an AGE-modified protein on a cell (“anti-AGE antibody” or “AGE antibody”) is known in the art. Examples include those described in U.S. 5,702,704 (Bucala) and U.S. 6,380,165 (Al-Abed et al.). Examples include an antibody that binds to one or more AGE-modified proteins having an AGE modification such as FFI, pyrraline, AFGP, ALI, carboxymethyllysine, carboxyethyllysine and pentosidine, and mixtures of such antibodies. Preferably, the antibody binds carboxymethyllysine-modified or carboxyethyllysine-modified proteins. Preferably, the antibody is non-immunogenic to the animal in which it will be used, such as non-immunogenic to humans; companion animals including cats, dogs and horses; and commercially important animals, such camels (or alpaca), cattle (bovine), sheep, and goats. More preferably, the antibody has the same species constant region as antibodies of the animal to reduce the immune response against the antibody, such as being humanized (for humans), felinized (for cats), caninized (for dogs), equuinized (for horses), carnalized (for camels or alpaca), bovinized (for cattle), ovinized (for sheep), or caperized (for goats). Most preferably, the antibody is identical to that of the animal in which it will be used (except for the variable region), such as a human antibody, a cat antibody, a dog antibody, a horse antibody, a camel antibody, a bovine antibody, a sheep antibody or a goat antibody. Details of the constant regions and other parts of antibodies for these animals are described below. The antibody may be monoclonal or polyclonal. Preferably, the antibody is a monoclonal antibody. [43] Particularly preferred anti-AGE antibodies include those which bind to proteins or peptides that exhibit a carboxymethyllysine or carboxyethyllysine AGE modification. Carboxymethyllysine (also known as N(epsilon)-(carboxymethyl)lysine, N(6)-carboxymethyllysine, or 2-Amino-6-(carboxymethylamino)hexanoic acid) and carboxyethyllysine (also known as N-epsilon-(carboxyethyl)lysine) are found on proteins or peptides and lipids as a result of oxidative stress and chemical glycation. CML- and CEL-modified proteins or peptides are recognized by the receptor RAGE which is expressed on a variety of cells. CML and CEL have been well-studied and CML- and CEL-related products are commercially available. For example, Cell Biolabs, Inc. sells CML-BSA antigens, CML polyclonal antibodies, CML immunoblot kits, and CML competitive ELISA kits (www.cellbiolabs.com/cml-assays) as well as CEL-BSA antigens and CEL competitive ELISA kits (www.cellbiolabs.com/cel-n- epsilon-carboxyethyl-lysine-assays-and-reagents). A particularly preferred antibody includes the variable region of the commercially available mouse anti-glycation endproduct antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin, the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247), modified to have a human constant region (or the constant region of the animal into which it will be administered). Commercially-available antibodies, such as the carboxymethyl lysine antibody corresponding to catalog no. MAB3247 from R&D Systems, Inc., may be intended for diagnostic purposes and may contain material that is not suited for use in animals or humans. Preferably, commercially-available antibodies are purified and/or isolated prior to use in animals or humans to remove toxins or other potentially-harmful material.
[44] The anti-AGE antibody has low rate of dissociation from the antibody-antigen complex, or kd (also referred to as kback or off-rate), preferably at most 9 x 10‘3, 8 x
10’3, 7 x 10’3 or 6 x 10’3 (sec1). The anti-AGE antibody has a high affinity for the AGE-modified protein of a cell, which may be expressed as a low dissociation constant KD of at most 9 x 10’6, 8 x 10’6, 7 x 10’6, 6 x 10’6, 5 x 10’6, 4 x 10-6 or 3 x 10'6 (M). Preferably, the binding properties of the anti-AGE antibody are similar to, the same as, or superior to the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247), illustrated in FIG. 1.
[45] The anti-AGE antibody may destroy AGE-modified cells through antibodydependent cell-mediated cytotoxicity (ADCC). ADCC is a mechanism of cell- mediated immune defense in which an effector cell of the immune system actively lyses a target cell whose membrane-surface antigens have been bound by specific antibodies. ADCC may be mediated by natural killer (NK) cells, macrophages, neutrophils or eosinophils. The effector cells bind to the Fc portion of the bound antibody. The anti-AGE antibody may also destroy AGE-modified cells through complement-dependent cytotoxicity (CDC). In CDC, the complement cascade of the immune system is triggered by an antibody binding to a target antigen.
[46] The anti-AGE antibody may be conjugated to an agent that causes the destruction of AGE-modified cells. Such agents may be a toxin, a cytotoxic agent, magnetic nanoparticles, and magnetic spin-vortex discs.
[47] A toxin, such as pore-forming toxins (PFT) (Aroian R. etal., “Pore-Forming Toxins and Cellular Non-lmmune Defenses (CNIDs),” Current Opinion in Microbiology, 10:57-61 (2007)), conjugated to an anti-AGE antibody may be injected into a patient to selectively target and remove AGE-modified cells. The anti-AGE antibody recognizes and binds to AGE-modified cells. Then, the toxin causes pore formation at the cell surface and subsequent cell removal through osmotic lysis.
[48] Magnetic nanoparticles conjugated to the anti-AGE antibody may be injected into a patient to target and remove AGE-modified cells. The magnetic nanoparticles can be heated by applying a magnetic field in order to selectively remove the AGE- modified cells.
[49] As an alternative, magnetic spin-vortex discs, which are magnetized only when a magnetic field is applied to avoid self-aggregation that can block blood vessels, begin to spin when a magnetic field is applied, causing membrane disruption of target cells. Magnetic spin-vortex discs, conjugated to anti-AGE antibodies specifically target AGE-modified cell types, without removing other cells. [50] Antibodies typically comprise two heavy chains and two light chains of polypeptides joined to form a "Y" shaped molecule. The constant region determines the mechanism used to target the antigen. The amino acid sequence in the tips of the "Y" (the variable region) varies among different antibodies. This variation gives the antibody its specificity for binding antigen. The variable region, which includes the ends of the light and heavy chains, is further subdivided into hypervariable (HV - also sometimes referred to as complementarity determining regions, or CDRs) and framework (FR) regions. When antibodies are prepared recombinantly, it is also possible to have a single antibody with variable regions (or complementary determining regions) that bind to two different antigens, with each tip of the “Y” being specific to each antigen; these are referred to as bi-specific antibodies.
[51] A humanized anti-AGE antibody according to the present invention may have the human constant region sequence of amino acids shown in SEQ ID NO: 22. The heavy chain complementarity determining regions of the humanized anti-AGE antibody may have one or more of the protein sequences shown in SEQ ID NO: 23 (CDR1H), SEQ ID NO: 24 (CDR2H) and SEQ ID NO: 25 (CDR3H). The light chain complementarity determining regions of the humanized anti-AGE antibody may have one or more of the protein sequences shown in SEQ ID NO: 26 (CDR1L), SEQ ID NO: 27 (CDR2L) and SEQ ID NO: 28 (CDR3L).
[52] The heavy chain of human Homo sapiens) antibody immunoglobulin G1 may have or may include the protein sequence of SEQ ID NO: 1. The variable domain of the heavy chain may have or may include the protein sequence of SEQ ID NO: 2. The complementarity determining regions of the variable domain of the heavy chain (SEQ ID NO: 2) are shown in SEQ ID NO: 41, SEQ ID NO: 42 and SEQ ID NO: 43. The kappa light chain of human (Homo sapiens) antibody immunoglobulin G1 may have or may include the protein sequence of SEQ ID NO: 3. The variable domain of the kappa light chain may have or may include the protein sequence of SEQ ID NO: 4. Optionally, the arginine (Arg or R) residue at position 128 of SEQ ID NO: 4 may be omitted. The complementarity determining regions of the variable domain of the light chain (SEQ ID NO: 4) are shown in SEQ ID NO: 44, SEQ ID NO: 45 and SEQ ID NO: 46. The variable regions may be codon-optimized, synthesized and cloned into expression vectors containing human immunoglobulin G1 constant regions. In addition, the variable regions may be used in the humanization of non-human antibodies.
[53] The antibody heavy chain may be encoded by the DNA sequence of SEQ ID NO: 12, a murine anti-AGE immunoglobulin G2b heavy chain. The protein sequence of the murine anti-AGE immunoglobulin G2b heavy chain encoded by SEQ ID NO: 12 is shown in SEQ ID NO: 16. The variable region of the murine antibody is shown in SEQ ID NO: 20, which corresponds to positions 25-142 of SEQ ID NO: 16. The antibody heavy chain may alternatively be encoded by the DNA sequence of SEQ ID NO: 13, a chimeric anti-AGE human immunoglobulin G1 heavy chain. The protein sequence of the chimeric anti-AGE human immunoglobulin G1 heavy chain encoded by SEQ ID NO: 13 is shown in SEQ ID NO: 17. The chimeric anti-AGE human immunoglobulin includes the murine variable region of SEQ ID NO: 20 in positions 25-142. The antibody light chain may be encoded by the DNA sequence of SEQ ID NO: 14, a murine anti-AGE kappa light chain. The protein sequence of the murine anti-AGE kappa light chain encoded by SEQ ID NO: 14 is shown in SEQ ID NO: 18. The variable region of the murine antibody is shown in SEQ ID NO: 21, which corresponds to positions 21-132 of SEQ ID NO: 18. The antibody light chain may alternatively be encoded by the DNA sequence of SEQ ID NO: 15, a chimeric anti- AGE human kappa light chain. The protein sequence of the chimeric anti-AGE human kappa light chain encoded by SEQ ID NO: 15 is shown in SEQ ID NO: 19. The chimeric anti-AGE human immunoglobulin includes the murine variable region of SEQ ID NO: 21 in positions 21-132.
[54] A humanized anti-AGE antibody according to the present invention may have or may include one or more humanized heavy chains or humanized light chains. A humanized heavy chain may be encoded by the DNA sequence of SEQ ID NO: 30, 32 or 34. The protein sequences of the humanized heavy chains encoded by SEQ ID NOs: 30, 32 and 34 are shown in SEQ ID NOs: 29, 31 and 33, respectively. A humanized light chain may be encoded by the DNA sequence of SEQ ID NO: 36, 38 or 40. The protein sequences of the humanized light chains encoded by SEQ ID NOs: 36, 38 and 40 are shown in SEQ ID NOs: 35, 37 and 39, respectively. Preferably, the humanized anti-AGE antibody maximizes the amount of human sequence while retaining the original antibody specificity. A complete humanized antibody may be constructed that contains a heavy chain having a protein sequence chosen from SEQ ID NOs: 29, 31 and 33 and a light chain having a protein sequence chosen from SEQ ID NOs: 35, 37 and 39.
[55] The protein sequence of an antibody from a non-human species may be modified to include the variable domain of the heavy chain having the sequence shown in SEQ ID NO: 2 or the kappa light chain having the sequence shown in SEQ ID NO: 4. The non-human species may be a companion animal, such as the domestic cat or domestic dog, or livestock, such as cattle, the horse or the camel. Preferably, the non-human species is not the mouse. The heavy chain of the horse (Equus caballus) antibody immunoglobulin gamma 4 may have or may include the protein sequence of SEQ ID NO: 5 (EMBL/GenBank accession number AY445518). The heavy chain of the horse (Equus caballus) antibody immunoglobulin delta may have or may include the protein sequence of SEQ ID NO: 6 (EMBL/GenBank accession number AY631942). The heavy chain of the dog (Canis familiaris) antibody immunoglobulin A may have or may include the protein sequence of SEQ ID NO: 7 (GenBank accession number L36871). The heavy chain of the dog (Canis familiaris) antibody immunoglobulin E may have or may include the protein sequence of SEQ ID NO: 8 (GenBank accession number L36872). The heavy chain of the cat (Felis catus) antibody immunoglobulin G2 may have or may include the protein sequence of SEQ ID NO: 9 (DDBJ/EMBL/GenBank accession number KF811175).
[56] Animals of the camelid family, such as camels (Camelus dromedarius and Camelus bactrianus), llamas (Lama glama, Lama pacos and Lama vicugna), alpacas (Vicugna pacos) and guanacos (Lama guanicoe), have a unique antibody that is not found in other mammals. In addition to conventional immunoglobulin G antibodies composed of heavy and light chain tetramers, camelids also have heavy chain immunoglobulin G antibodies that do not contain light chains and exist as heavy chain dimers. These antibodies are known as heavy chain antibodies, HCAbs, single-domain antibodies or sdAbs, and the variable domain of a camelid heavy chain antibody is known as the VHH. The camelid heavy chain antibodies lack the heavy chain CH1 domain and have a hinge region that is not found in other species. The variable region of the Arabian camel (Camelus dromedarius) single-domain antibody may have or may include the protein sequence of SEQ ID NO: 10 (GenBank accession number AJ245148). The variable region of the heavy chain of the Arabian camel (Camelus dromedarius) tetrameric immunoglobulin may have or may include the protein sequence of SEQ ID NO: 11 (GenBank accession number AJ245184).
[57] In addition to camelids, heavy chain antibodies are also found in cartilaginous fishes, such as sharks, skates and rays. This type of antibody is known as an immunoglobulin new antigen receptor or IgNAR, and the variable domain of an IgNAR is known as the VNAR. The IgNAR exists as two identical heavy chain dimers composed of one variable domain and five constant domains each. Like camelids, there is no light chain.
[58] The protein sequences of additional non-human species may be readily found in online databases, such as the International ImMunoGeneTics Information System (www.imgt.org), the European Bioinformatics Institute (www.ebi.ac.uk), the DNA Databank of Japan (ddbj.nig.ac.jp/arsa) or the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov).
[59] An anti-AGE antibody or a variant thereof may include a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 20, including post-translational modifications thereof. A variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity may contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-AGE antibody including that sequence retains the ability to bind to AGE. The substitutions, insertions, or deletions may occur in regions outside the variable region.
[60] An anti-AGE antibody or a variant thereof may include a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 21 , including post-translational modifications thereof. A variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity may contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-AGE antibody including that sequence retains the ability to bind to AGE. The substitutions, insertions, or deletions may occur in regions outside the variable region.
[61] Alternatively, the antibody may have the complementarity determining regions of commercially available mouse anti-glycation end-product antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin (CML-KLH), the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247).
[62] The antibody may have or may include constant regions which permit destruction of targeted cells by a subject’s immune system.
[63] Mixtures of antibodies that bind to more than one type AGE of AGE-modified proteins may also be used.
[64] Bi-specific antibodies, which are anti-AGE antibodies directed to two different epitopes, may also be used. Such antibodies will have a variable region (or complementary determining region) from those of one anti-AGE antibody, and a variable region (or complementary determining region) from a different antibody.
[65] Antibody fragments may be used in place of whole antibodies. For example, immunoglobulin G may be broken down into smaller fragments by digestion with enzymes. Papain digestion cleaves the N-terminal side of inter-heavy chain disulfide bridges to produce Fab fragments. Fab fragments include the light chain and one of the two N-terminal domains of the heavy chain (also known as the Fd fragment). Pepsin digestion cleaves the C-terminal side of the inter-heavy chain disulfide bridges to produce F(ab’)2 fragments. F(ab’)2 fragments include both light chains and the two N-terminal domains linked by disulfide bridges. Pepsin digestion may also form the Fv (fragment variable) and Fc (fragment crystallizable) fragments. The Fv fragment contains the two N-terminal variable domains. The Fc fragment contains the domains which interact with immunoglobulin receptors on cells and with the initial elements of the complement cascade. Pepsin may also cleave immunoglobulin G before the third constant domain of the heavy chain (CH3) to produce a large fragment F(abc) and a small fragment pFc’. Antibody fragments may alternatively be produced recombinantly. Preferably, such antibody fragments are conjugated to an agent that causes the destruction of AGE-modified cells.
[66] If additional antibodies are desired, they can be produced using well-known methods. For example, polyclonal antibodies (pAbs) can be raised in a mammalian host by one or more injections of an immunogen, and if desired, an adjuvant. Typically, the immunogen (and adjuvant) is injected in a mammal by a subcutaneous or intraperitoneal injection. The immunogen may be an AGE-modified protein of a cell, such as AGE-antithrombin III, AGE-calmodulin, AGE-insulin, AGE- ceruloplasmin, AGE-collagen, AGE-cathepsin B, AGE-albumin such as AGE-bovine serum albumin (AGE-BSA), AGE-human serum albumin and ovalbumin, AGE- crystallin, AGE-plasminogen activator, AGE-endothelial plasma membrane protein, AGE-aldehyde reductase, AGE-transferrin, AGE-fibrin, AGE-copper/zinc SOD, AGE- apo B, AGE-fibronectin, AGE-pancreatic ribose, AGE-apo A-l and II, AGE- hemoglobin, AGE-Na+/K+-ATPase, AGE-plasminogen, AGE-myelin, AGE-lysozyme, AGE-immunoglobulin, AGE-red cell Glu transport protein, AGE-p-N-acetyl hexominase, AGE-apo E, AGE-red cell membrane protein, AGE-aldose reductase, AGE-ferritin, AGE-red cell spectrin, AGE-alcohol dehydrogenase, AGE-haptoglobin, AGE-tubulin, AGE-thyroid hormone, AGE-fibrinogen, AGE- -microglobulin, AGE- sorbitol dehydrogenase, AGE-ai-antitrypsin, AGE-carbonate dehydratase, AGE- RNAse, AG E-low density lipoprotein, AGE-hexokinase, AGE-apo C-l, AGE-RNAse, AGE-hemoglobin such as AGE-human hemoglobin, AGE-low density lipoprotein (AGE-LDL) and AGE-collagen IV. AGE-modified cells, such as AGE-modified erythrocytes, whole, lysed, or partially digested, may also be used as AGE antigens. Examples of adjuvants include Freund’s complete, monophosphoryl Lipid A synthetic-trehalose dicorynomycolate, aluminum hydroxide (alum), heat shock proteins HSP 70 or HSP96, squalene emulsion containing monophosphoryl lipid A, a2-macroglobulin and surface active substances, including oil emulsions, pleuronic polyols, polyanions and dinitrophenol. To improve the immune response, an immunogen may be conjugated to a polypeptide that is immunogenic in the host, such as keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, cholera toxin, labile enterotoxin, silica particles or soybean trypsin inhibitor. A preferred immunogen conjugate is AGE-KLH. Alternatively, pAbs may be made in chickens, producing IgY molecules.
[67] Monoclonal antibodies (mAbs) may also be made by immunizing a host or lymphocytes from a host, harvesting the mAb-secreting (or potentially secreting) lymphocytes, fusing those lymphocytes to immortalized cells (for example, myeloma cells), and selecting those cells that secrete the desired mAb. Other techniques may be used, such as the EBV-hybridoma technique. Techniques for the generation of chimeric antibodies by splicing genes encoding the variable domains of antibodies to genes of the constant domains of human (or other animal) immunoglobulin result in "chimeric antibodies" that are substantially human (humanized) or substantially “ized” to another animal (such as cat, dog, horse, camel or alpaca, cattle, sheep, or goat) at the amino acid level. If desired, the mAbs may be purified from the culture medium or ascites fluid by conventional procedures, such as protein A-sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, ammonium sulfate precipitation or affinity chromatography. Additionally, human monoclonal antibodies can be generated by immunization of transgenic mice containing a third copy IgG human trans-loci and silenced endogenous mouse Ig loci or using human-transgenic mice. Production of humanized monoclonal antibodies and fragments thereof can also be generated through phage display technologies.
[68] A "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Preferred examples of such carriers or diluents include water, saline, Ringer’s solutions and dextrose solution. Supplementary active compounds can also be incorporated into the compositions. Solutions and suspensions used for parenteral administration can include a sterile diluent, such as water for injection, saline solution, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[69] The antibodies may be administered by injection, such as by intravenous injection or intra-articular injection into an affected joint. Pharmaceutical compositions suitable for injection include sterile aqueous solutions or dispersions for the extemporaneous preparation of sterile injectable solutions or dispersion. Various excipients may be included in pharmaceutical compositions of antibodies suitable for injection. Suitable carriers include physiological saline, bacteriostatic water, CREMOPHOR EL® (BASF; Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid so as to be administered using a syringe. Such compositions should be stable during manufacture and storage and must be preserved against contamination from microorganisms such as bacteria and fungi. Various antibacterial and anti-fungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal, can contain microorganism contamination. Isotonic agents such as sugars, polyalcohols, such as manitol, sorbitol, and sodium chloride can be included in the composition. Compositions that can delay absorption include agents such as aluminum monostearate and gelatin. Sterile injectable solutions can be prepared by incorporating antibodies, and optionally other therapeutic components, in the required amount in an appropriate solvent with one or a combination of ingredients as required, followed by sterilization. Methods of preparation of sterile solids for the preparation of sterile injectable solutions include vacuum drying and freeze-drying to yield a solid.
[70] For administration by inhalation, the antibodies may be delivered as an aerosol spray from a nebulizer or a pressurized container that contains a suitable propellant, for example, a gas such as carbon dioxide. Antibodies may also be delivered via inhalation as a dry powder, for example using the iSPERSE™ inhaled drug delivery platform (PULMATRIX, Lexington, Mass.). The use of anti-AGE antibodies which are chicken antibodies (IgY) may be non-immunogenic in a variety of animals, including humans, when administered by inhalation.
[71] An appropriate dosage level of each type of antibody will generally be about
0.01 to 500 mg per kg patient body weight Preferably, the dosage level will be about 0.1 to about 250 mg/kg; more preferably about 0.5 to about 100 mg/kg. A suitable dosage level may be about 0.01 to 250 mg/kg, about 0.05 to 100 mg/kg, or about 0.1 to 50 mg/kg. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg. Although each type of antibody may be administered on a regimen of 1 to 4 times per day, such as once or twice per day, antibodies typically have a long half-life in vivo. Accordingly, each type of antibody may be administered once a day, once a week, once every two or three weeks, once a month, or once every 60 to 90 days.
[72] A subject that receives administration of an anti-AGE antibody may be tested to determine if the administration has been effective to treat osteoarthritis by monitoring symptoms of osteoarthritis overtime, such as pain level, stiffness and joint functionality. These factors may be measured directly or may be analyzed according to a standard medical grading scale, such as the WOMAC scale. Osteoarthritis may also be evaluated radiographically, such as by analysis of X-rays or magnetic resonance imaging (MRI) radiographs. Diagnostic imaging can reveal signs of osteoarthritis including joint space narrowing, increased subchondral bone density, increased subchondral stenosis (increased bone formation), subchondral cyst formation, osteophyte formation and hard tissue enlargement. A subject may be considered to have received an effective antibody treatment if he or she demonstrates an improvement in osteoarthritis symptoms between subsequent measurements or over time. Alternatively, the concentration and/or number of senescent cells may be measured overtime. Administration of antibody and subsequent testing may be repeated until the desired therapeutic result is achieved. [73] Unit dosage forms can be created to facilitate administration and dosage uniformity. Unit dosage form refers to physically discrete units suited as single dosages for the subject to be treated, containing a therapeutically effective quantity of one or more types of antibodies in association with the required pharmaceutical carrier. Preferably, the unit dosage form is in a sealed container and is sterile.
[74] Vaccines against AGE-modified proteins or peptides contain an AGE antigen, an adjuvant, optional preservatives and optional excipients. Examples of AGE antigens include AGE-modified proteins or peptides such as AGE-antithrombin III, AGE-calmodulin, AGE-insulin, AGE-ceruloplasmin, AGE-collagen, AGE-cathepsin B, AGE-albumin such as AGE-bovine serum albumin (AGE-BSA), AGE-human serum albumin and ovalbumin, AGE-crystallin, AGE-plasminogen activator, AGE- endothelial plasma membrane protein, AGE-aldehyde reductase, AGE-transferrin, AGE-fibrin, AGE-copper/zinc SOD, AGE-apo B, AGE-fibronectin, AGE-pancreatic ribose, AGE-apo A-l and II, AGE-hemoglobin, AGE-Na+/K+-ATPase, AGE- plasminogen, AGE-myelin, AGE-lysozyme, AGE-immunoglobulin, AGE-red cell Glu transport protein, AGE- -N-acetyl hexominase, AGE-apo E, AGE-red cell membrane protein, AGE-aldose reductase, AGE-ferritin, AGE-red cell spectrin, AGE-alcohol dehydrogenase, AGE-haptoglobin, AGE-tubulin, AGE-thyroid hormone, AGE- fibrinogen, AGE-P2-microglobulin, AGE-sorbitol dehydrogenase, AGE-ai-antitrypsin, AGE-carbonate dehydratase, AGE-RNAse, AGE-low density lipoprotein, AGE- hexokinase, AGE-apo C-l , AGE-RNAse, AGE-hemoglobin such as AGE-human hemoglobin, AGE-low density lipoprotein (AGE-LDL) and AGE-collagen IV. AGE- modified cells, such as AGE-modified erythrocytes, whole, lysed, or partially digested, may also be used as AGE antigens. Suitable AGE antigens also include proteins or peptides that exhibit AGE modifications (also referred to as AGE epitopes or AGE moieties) such as carboxymethyllysine (CML), carboxyethyllysine (CEL), pentosidine, pyrraline, FFI, AFGP and ALL The AGE antigen may be an AGE- protein conjugate, such as AGE conjugated to keyhole limpet hemocyanin (AGE- KLH). Further details of some of these AGE-modified proteins or peptides and their preparation are described in Bucala. [75] Particularly preferred AGE antigens include proteins or peptides that exhibit a carboxymethyllysine or carboxyethyllysine AGE modification. Carboxymethyllysine (also known as N(epsilon)-(carboxymethyl)lysine, N(6)-carboxymethyllysine, or 2- Amino-6-(carboxymethylamino)hexanoic acid) and carboxyethyllysine (also known as N-epsilon-(carboxyethyl)lysine) are found on proteins or peptides and lipids as a result of oxidative stress and chemical glycation, and have been correlated with juvenile genetic disorders. CML- and CEL-modified proteins or peptides are recognized by the receptor RAGE which is expressed on a variety of cells. CML and CEL have been well-studied and CML- and CEL-related products are commercially available. For example, Cell Biolabs, Inc. sells CM L-BSA antigens, CML polyclonal antibodies, CML immunoblot kits, and CML competitive ELISA kits (www.cellbiolabs.com/cml-assays) as well as CEL-BSA antigens and CEL competitive ELISA kits (www.cellbiolabs.com/cel-n-epsilon-carboxyethyl-lysine- assays-and-reagents).
[76] AGE antigens may be conjugated to carrier proteins to enhance antibody production in a subject. Antigens that are not sufficiently immunogenic alone may require a suitable carrier protein to stimulate a response from the immune system. Examples of suitable carrier proteins include keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, cholera toxin, labile enterotoxin, silica particles and soybean trypsin inhibitor. Preferably, the carrier protein is KLH (AGE-KLH). KLH has been extensively studied and has been identified as an effective carrier protein in experimental cancer vaccines. Preferred AGE antigen-carrier protein conjugates include CML-KLH and CEL-KLH.
[77] The administration of an AGE antigen allows the immune system to develop immunity to the antigen. Immunity is a long-term immune response, either cellular or humoral. A cellular immune response is activated when an antigen is presented, preferably with a co-stimulator to a T-cell which causes it to differentiate and produce cytokines. The cells involved in the generation of the cellular immune response are two classes of T-helper (Th) cells, Th1 and Th2. Th1 cells stimulate B cells to produce predominantly antibodies of the lgG2A isotype, which activates the complement cascade and binds the Fc receptors of macrophages, while Th2 cells stimulate B cells to produce lgG1 isotype antibodies in mice, lgG4 isotype antibodies in humans, and IgE isotype antibodies. The human body also contains “professional” antigen-presenting cells such as dendritic cells, macrophages, and B cells.
[78] A humoral immune response is triggered when a B cell selectively binds to an antigen and begins to proliferate, leading to the production of a clonal population of cells that produce antibodies that specifically recognize that antigen and which may differentiate into antibody-secreting cells, referred to as plasma-cells or memory-B cells. Antibodies are molecules produced by B-cells that bind a specific antigen. The antigen-antibody complex triggers several responses, either cell-mediated, for example by natural killers (NK) or macrophages, or serum-mediated, for example by activating the complement system, a complex of several serum proteins that act sequentially in a cascade that result in the lysis of the target cell.
[79] Immunological adjuvants (also referred to simply as “adjuvants”) are the component(s) of a vaccine which augment the immune response to the immunogenic agent Adjuvants function by attracting macrophages to the immunogenic agent and then presenting the agent to the regional lymph nodes to initiate an effective antigenic response. Adjuvants may also act as carriers themselves for the immunogenic agent. Adjuvants may induce an inflammatory response, which may play an important role in initiating the immune response.
[80] Adjuvants include mineral compounds such as aluminum salts, oil emulsions, bacterial products, liposomes, immunostimulating complexes and squalene.
Aluminum compounds are the most widely used adjuvants in human and veterinary vaccines. These aluminum compounds include aluminum salts such as aluminum phosphate (AIPO4) and aluminum hydroxide (AI(OH)3) compounds, typically in the form of gels, and are generically referred to in the field of vaccine immunological adjuvants as "alum." Aluminum hydroxide is a poorly crystalline aluminum oxyhydroxide having the structure of the mineral boehmite. Aluminum phosphate is an amorphous aluminum hydroxy phosphate. Negatively charged species (for example, negatively charged antigens) can absorb onto aluminum hydroxide gels at neutral pH, whereas positively charged species (for example, positively charged antigens) can absorb onto aluminum phosphate gels at neutral pH. It is believed that these aluminum compounds provide a depot of antigen at the site of administration, thereby providing a gradual and continuous release of antigen to stimulate antibody production. Aluminum compounds tend to more effectively stimulate a cellular response mediated by Th2, rather than Th1 cells.
[81] Emulsion adjuvants include water-in-oil emulsions (for example, Freund's adjuvants, such as killed mycobacteria in oil emulsion) and oil-in-water emulsions (for example, MF-59). Emulsion adjuvants include an immunogenic component, for example squalene (MF-59) or mannide oleate (Incomplete Freund's Adjuvants), which can induce an elevated humoral response, increased T cell proliferation, cytotoxic lymphocytes and cell-mediated immunity.
[82] Liposomal or vesicular adjuvants (including paucilamellar lipid vesicles) have lipophilic bilayer domains and an aqueous milieu which can be used to encapsulate and transport a variety of materials, for example an antigen. Paucilamellar vesicles (for example, those described in U.S. Pat. No. 6,387,373) can be prepared by mixing, under high pressure or shear conditions, a lipid phase comprising a nonphospholipid material (for example, an amphiphile surfactant; see U.S. Pat Nos. 4,217,344; 4,917,951; and 4,911,928), optionally a sterol, and any water-immiscible oily material to be encapsulated in the vesicles (for example, an oil such as squalene oil and an oil-soluble or oil-suspended antigen); and an aqueous phase such as water, saline, buffer or any other aqueous solution used to hydrate the lipids. Liposomal or vesicular adjuvants are believed to promote contact of the antigen with immune cells, for example by fusion of the vesicle to the immune cell membrane, and preferentially stimulate the Th1 sub-population of T-helper cells.
[83] Other types of adjuvants include Mycobacterium bovis bacillus Calmette- Guerin (BCG), quill-saponin and unmethylated CpG dinucleotides (CpG motifs). Additional adjuvants are described in U.S. Patent Application Publication Pub. No. US 2010/0226932 (September 9, 2010) and Jiang, Z-H. et al. “Synthetic vaccines: the role of adjuvants in immune targeting”, Current Medicinal Chemistry, Vol. 10(15), pp. 1423-39 (2003). Preferable adjuvants include Freund’s complete adjuvant and Freund’s incomplete adjuvant.
[84] The vaccine may optionally include one or more preservatives, such as antioxidants, antibacterial and antimicrobial agents, as well as combinations thereof. Examples include benzethonium chloride, ethylenediamine-tetraacetic acid sodium (EDTA), thimerosal, phenol, 2-phenoxyethanol, formaldehyde and formalin; antibacterial agents such as amphotericin B, chlortetracycline, gentamicin, neomycin, polymyxin B and streptomycin; antimicrobial surfactants such as polyoxyethylene-9, 10-nonyl phenol (Triton N-101 , octoxynol-9), sodium deoxycholate and polyoxyethylated octyl phenol (Triton X-I00). The production and packaging of the vaccine may eliminate the need for a preservative. For example, a vaccine that has been sterilized and stored in a sealed container may not require a preservative.
[85] Other components of vaccines include pharmaceutically acceptable excipients, such as stabilizers, thickening agents, toxin detoxifiers, diluents, pH adjusters, tonicity adjustors, surfactants, antifoaming agents, protein stabilizers, dyes and solvents. Examples of such excipients include hydrochloric acid, phosphate buffers, sodium acetate, sodium bicarbonate, sodium borate, sodium citrate, sodium hydroxide, potassium chloride, potassium chloride, sodium chloride, polydimethylsilozone, brilliant green, phenol red (phenolsulfon-phthalein), glycine, glycerin, sorbitol, histidine, monosodium glutamate, potassium glutamate, sucrose, urea, lactose, gelatin, sorbitol, polysorbate 20, polysorbate 80 and glutaraldehyde. A variety of these components of vaccines, as well as adjuvants, are described in www.cdc.gov/vaccines/pubs/pinkbook/downloads/appendices/BZexcipient-table-2.pdf and Vogel, F. R. et al., “A compendium of vaccine adjuvants and excipients”, Pharmaceutical Biotechnology, Vol. 6, pp. 141-228 (1995).
[86] The vaccine may contain from 1 pg to 100 mg of at least one AGE antigen, including 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 400, 800 or 1000 pg, or 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80 or 90 mg. The amount used for a single injection corresponds to a unit dosage. [87] The vaccine may be provided in unit dosage form or in multidosage form, such as 2-100 or 2-10 doses. The unit dosages may be provided in a vial with a septum, or in a syringe with or without a needle. The vaccine may be administered intravenously, subdermally or intraperitoneally. Preferably, the vaccine is sterile.
[88] The vaccine may be administered one or more times, such as 1 to 10 times, including 2, 3, 4, 5, 6, 7, 8 or 9 times, and may be administered over a period of time ranging from 1 week to 1 year, 2-10 weeks or 2-10 months. Furthermore, booster vaccinations may be desirable, over the course of 1 year to 20 years, including 2, 5, 10 and 15 years.
[89] A subject that receives a vaccine for AGE-modified proteins or peptides of a cell may be tested to determine if he or she has developed an immunity to the AGE- modified proteins or peptides. Suitable tests may include blood tests for detecting the presence of an antibody, such as immunoassays or antibody titers. Alternatively, an immunity to AGE-modified proteins or peptides may be determined by monitoring osteoarthritis symptoms of a subject over time. For example, a baseline knee stiffness of a subject may be measured followed by administration of the vaccine for AGE-modified proteins or peptides of a cell. Immunity to AGE-modified proteins or peptides may be determined by periodically measuring the knee stiffness of the subject and comparing the subsequent measurements to the baseline measurement. A subject may be considered to have developed an immunity to AGE-modified proteins or peptides if he or she demonstrates an improvement in knee stiffness between subsequent measurements or over time. An immunity to AGE-modified proteins or peptides may also be determined by monitoring the concentration and/or number of senescent cells over time. In addition to testing for the development of an immunity to AGE-modified proteins or peptides, a subject may also be tested to determine if the vaccination has been effective to treat osteoarthritis. The effectiveness of the vaccination may be determined by vaccinating a subject followed by periodically obtaining X-rays and/or MRI images of the subject over time or measuring the concentration and/or number of senescent cells. Vaccination and subsequent testing may be repeated until the desired therapeutic result is achieved. [90] The vaccination process may be designed to provide immunity against multiple AGE moieties. A single AGE antigen may induce the production of AGE antibodies which are capable of binding to multiple AGE moieties. Alternatively, the vaccine may contain multiple AGE antigens. In addition, a subject may receive multiple vaccines, where each vaccine contains a different AGE antigen.
[91] Any mammal that could develop osteoarthritis may be treated by the methods herein described. Humans are a preferred mammal for treatment. Other mammals that may be treated include mice, rats, goats, sheep, cows, horses and companion animals, such as dogs or cats. Alternatively, any of the mammals or subjects identified above may be excluded from the patient population in need of treatment for osteoarthritis.
[92] A subject may be identified as having osteoarthritis or in need of treatment if he or she exhibits any of the classical osteoarthritis symptoms, such as joint pain, stiffness, reduced range of motion, joint swelling or weakness or numbness of the arms and legs. A subject may also be identified as in need of treatment based on analysis of diagnostic images such as X-rays and MRI radiographs for the presence of joint space narrowing, increased subchondral bone density, increased subchondral stenosis (increased bone formation), subchondral cyst formation, osteophyte formation or hard tissue enlargement. In addition, subjects may be identified as in need of treatment due to specific physical changes that are known risk factors for osteoarthritis such as a direct injury to a joint or having legs of unequal length. Subjects may also be identified as in need of treatment based on the presence of a pathological condition associated with AGEs such as, for example, atherosclerosis, inflammation, retinopathy, nephropathy, stroke, endothelial cell dysfunction, neurodegenerative disorders or cancer.
[93] The one-letter amino acid sequence that corresponds to SEQ ID NO: 1 is shown below:
10 20 30 40 50
MNLLLILTFV AAAVAQVQLL QPGAELVKPG ASVKLACKAS GYLFTTYWMH 60 70 80 90
WLKQRPGQGL EWIGEISPTN GRAYYNARFK SEATLTVDKS 100 110 120 130
SNTAYMQLSS LTSEASAVYY CARAYGNYEF AYWGQGTLVT 140 150 160 170
VSVASTKGPS VFPLAPSSKS TSGGTAALGC LVKDYFPEPV 180 190 200 210 220
TVSWNSGALT SGVHTFPAVL QSSGLYSLSS WTVPSSSLG TQTYICNVNH 230 240 250 260
KPSNTKVDKK VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP 270 280 290 300
PKPKDTLMIS RTPEVTCVW DVSHEDPEVK FNWYVDGVEV
310 320 330 340
HNAKTKPREE QYNSTYRWS VLTVLHQDWL NGKEYKCKVS
350 360 370 380 390
NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS LTCLVKGFYP
400 410 420 430
SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK 440 450 460
SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK
[94] Positions 16-133 of the above amino acid sequence correspond to SEQ ID NO: 2. Positions 46-50 of the above amino acid sequence correspond to SEQ ID NO: 41. Positions 65-81 of the above amino acid sequence correspond to SEQ ID NO: 42. Positions 114-122 of the above amino acid sequence correspond to SEQ ID NO: 43.
[95] The one-letter amino acid sequence that corresponds to SEQ ID NO: 3 is shown below:
10 20 30 40 50
MNLLLILTFV AAAVADWMT QTPLSLPVSL GDQASISCRS RQSLVNSNGN 60 70 80 90 100
TFLQWYLQKP GQSPKLLIYK VSLRFSGVPD RFSGSGSGTD FTLKISRVEA 110 120 130 140 150
EDLGLYFCSQ STHVPPTFGG GTKLEIKRTV AAPSVFIFPP SDEQLKSGTA 160 170 180 190
SWCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD 200 210 220 230
STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC
[96] Positions 16-128 of the above amino acid sequence correspond to SEQ ID NO: 4. Optionally, the arginine (Arg or R) residue at position 128 of SEQ ID NO: 4 may be omitted. Positions 39-54 of the above amino acid sequence correspond to SEQ ID NO: 44. Positions 70-76 of the above amino acid sequence correspond to
SEQ ID NO: 45. Positions 109-117 of the above amino acid sequence correspond to
SEQ ID NO: 46.
[97] The DNA sequence that corresponds to SEQ ID NO: 12 is shown below:
ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC
CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGCTGCAGCCAGGTGCCGAGCTC
GTGAAACCTGGCGCCTCTGTGAAGCTGGCCTGCAAGGCTTCCGGCTACCTGTT
CACCACCTACTGGATGCACTGGCTGAAGCAGAGGCCAGGCCAGGGCCTGGAA
TGGATCGGCGAGATCTCCCCCACCAACGGCAGAGCCTACTACAACGCCCGGTT
CAAGTCCGAGGCCACCCTGACCGTGGACAAGTCCTCCAACACCGCCTACATGC
AGCTGTCCTCCCTGACCTCTGAGGCCTCCGCCGTGTACTACTGCGCCAGAGCT
TACGGCAACTACGAGTTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTC
TGTGGCTAAGACCACCCCTCCCTCCGTGTACCCTCTGGCTCCTGGCTGTGGCG
ACACCACCGGATCCTCTGTGACCCTGGGCTGCCTCGTGAAGGGCTACTTCCCT
GAGTCCGTGACCGTGACCTGGAACTCCGGCTCCCTGTCCTCCTCCGTGCACAC
CTTTCCAGCCCTGCTGCAGTCCGGCCTGTACACCATGTCCTCCAGCGTGACAG
TGCCCTCCTCCACCTGGCCTTCCCAGACCGTGACATGCTCTGTGGCCCACCCT
GCCTCTTCCACCACCGTGGACAAGAAGCTGGAACCCTCCGGCCCCATCTCCAC
CATCAACCCTTGCCCTCCCTGCAAAGAATGCCACAAGTGCCCTGCCCCCAACC TGGAAGGCGGCCCTTCCGTGTTCATCTTCCCACCCAACATCAAGGACGTGCTG ATGATCTCCCTGACCCCCAAAGTGACCTGCGTGGTGGTGGACGTGTCCGAGGA CGACCCTGACGTGCAGATCAGTTGGTTCGTGAACAACGTGGAAGTGCACACCG CCCAGACCCAGACACACAGAGAGGACTACAACAGCACCATCAGAGTGGTGTCT ACCCTGCCCATCCAGCACCAGGACTGGATGTCCGGCAAAGAATTCAAGTGCAA AGTGAACAACAAGGACCTGCCCAGCCCCATCGAGCGGACCATCTCCAAGATCA AGGGCCTCGTGCGGGCTCCCCAGGTGTACATTCTGCCTCCACCAGCCGAGCA
GCTGTCCCGGAAGGATGTGTCTCTGACATGTCTGGTCGTGGGCTTCAACCCCG GCGACATCTCCGTGGAATGGACCTCCAACGGCCACACCGAGGAAAACTACAAG GACACCGCCCCTGTGCTGGACTCCGACGGCTCCTACTTCATCTACTCCAAGCT GAACATGAAGACCTCCAAGTGGGAAAAGACCGACTCCTTCTCCTGCAACGTGC GGCACGAGGGCCTGAAGAACTACTACCTGAAGAAAACCATCTCCCGGTCCCCC GGCTAG
[98] The DNA sequence that corresponds to SEQ ID NO: 13 is shown below:
ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGCTGCAGCCAGGTGCCGAGCTC GTGAAACCTGGCGCCTCTGTGAAGCTGGCCTGCAAGGCTTCCGGCTACCTGTT CACCACCTACTGGATGCACTGGCTGAAGCAGAGGCCAGGCCAGGGCCTGGAA TGGATCGGCGAGATCTCCCCCACCAACGGCAGAGCCTACTACAACGCCCGGTT CAAGTCCGAGGCCACCCTGACCGTGGACAAGTCCTCCAACACCGCCTACATGC AGCTGTCCTCCCTGACCTCTGAGGCCTCCGCCGTGTACTACTGCGCCAGAGCT
TACGGCAACTACGAGTTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTC TGTGGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCCCAGCAGCAAG AGCACCAGCGGCGGAACCGCCGCCCTGGGCTGCCTGGTGAAGGACTACTTCC CCGAGCCCGTGACCGTGTCCTGGAACAGCGGCGCTCTGACCAGCGGAGTGCA CACCTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTG GTGACCGTGCCCAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAA CCACAAGCCCTCCAACACCAAGGTGGACAAGAAGGTGGAGCCTAAGAGCTGC
GACAAGACCCACACCTGCCCTCCCTGCCCCGCCCCCGAGCTGCTGGGCGGAC CCAGCGTGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGC ACCCCCGAGGTGACCTGCGTGGTGGTGGACGTGAGCCACGAGGACCCCGAGG TGAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCACAACGCCAAGACCAAG CCTCGGGAGGAGCAGTACAACTCCACCTACCGCGTGGTGAGCGTGCTGACCG TGCTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTGAGCAA CAAGGCCCTGCCCGCTCCCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAG CCCCGGGAGCCTCAGGTGTACACCCTGCCCCCCAGCCGCGACGAGCTGACCA AGAACCAGGTGAGCCTGACCTGCCTGGTGAAGGGCTTCTACCCCTCCGACATC GCCGTGGAGTGGGAGAGCAACGGCCAGCCTGAGAACAACTACAAGACCACCC CTCCCGTGCTGGACAGCGACGGCAGCTTCTTCCTGTACAGCAAGCTGACCGTG GACAAGTCCCGGTGGCAGCAGGGCAACGTGTTCAGCTGCAGCGTGATGCACG AGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCCCGGATA G
[99] The DNA sequence that corresponds to SEQ ID NO: 14 is shown below:
ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGACCCCTCTGTCCCTGCCTGTGTCTCTG GGCGACCAGGCCTCCATCTCCTGCCGGTCTAGACAGTCCCTCGTGAACTCCAA CGGCAACACCTTCCTGCAGTGGTATCTGCAGAAGCCCGGCCAGTCCCCCAAGC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACAGATTTTCC GGCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGA GGACCTGGGCCTGTACTTCTGCAGCCAGTCCACCCACGTGCCCCCTACATTTG GCGGAGGCACCAAGCTGGAAATCAAACGGGCAGATGCTGCACCAACTGTATCC ATCTTCCCACCATCCAGTGAGCAGTTAACATCTGGAGGTGCCTCAGTCGTGTGC TTCTTGAACAACTTCTACCCCAAAGACATCAATGTCAAGTGGAAGATTGATGGC AGTGAACGACAAAATGGCGTCCTGAACAGTTGGACTGATCAGGACAGCAAAGA CAGCACCTACAGCATGAGCAGCACCCTCACGTTGACCAAGGACGAGTATGAAC GACATAACAGCTATACCTGTGAGGCCACTCACAAGACATCAACTTCACCCATTG TCAAGAGCTTCAACAGGAATGAGTGTTGA
[100] The DNA sequence that corresponds to SEQ ID NO: 15 is shown below:
ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT
CCACCGGAGACGTCGTGATGACCCAGACCCCTCTGTCCCTGCCTGTGTCTCTG GGCGACCAGGCCTCCATCTCCTGCCGGTCTAGACAGTCCCTCGTGAACTCCAA CGGCAACACCTTCCTGCAGTGGTATCTGCAGAAGCCCGGCCAGTCCCCCAAGC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACAGATTTTCC GGCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGA GGACCTGGGCCTGTACTTCTGCAGCCAGTCCACCCACGTGCCCCCTACATTTG GCGGAGGCACCAAGCTGGAAATCAAGCGGACCGTGGCCGCCCCCAGCGTGTT CATCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGT GCCTGCTGAACAACTTCTACCCCCGCGAGGCCAAGGTGCAGTGGAAGGTGGA CAACGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGACTCC AAGGACAGCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTA CGAGAAGCACAAGGTGTACGCCTGCGAGGTGACCCACCAGGGACTGTCTAGC CCCGTGACCAAGAGCTTCAACCGGGGCGAGTGCTAA
[101] The one-letter amino acid sequence that corresponds to SEQ ID NO: 16 is shown below:
MDPKGSLSWRILLFLSLAFELSYGQVQLLQPGAELVKPGASVKLACKASGYLFTTY WMHWLKQRPGQGLEWIGEISPTNGRAYYNARFKSEATLTVDKSSNTAYMQLSSLT SEASAVYYCARAYGNYEFAYWGQGTLVTVSVAKTTPPSVYPLAPGCGDTTGSSVT LGCLVKGYFPESVTVTWNSGSLSSSVHTFPALLQSGLYTMSSSVTVPSSTWPSQT VTCSVAHPASSTTVDKKLEPSGPISTINPCPPCKECHKCPAPNLEGGPSVFIFPPNIK DVLMISLTPKVTCVWDVSEDDPDVQISWFVNNVEVHTAQTQTHREDYNSTIRVVS TLPIQHQDWMSGKEFKCKVNNKDLPSPIERTISKIKGLVRAPQVYILPPPAEQLSRK DVSLTCLWGFNPGDISVEWTSNGHTEENYKDTAPVLDSDGSYFIYSKLNMKTSKW EKTDSFSCNVRHEGLKNYYLKKTISRSPG*
[102] The alanine residue at position 123 of the above amino acid sequence may optionally be replaced with a serine residue. The tyrosine residue at position 124 of the above amino acid sequence may optionally be replaced with a phenylalanine residue. Positions 25-142 of the above amino acid sequence correspond to SEQ ID NO: 20. SEQ ID NO: 20 may optionally include the substitutions at positions 123 and 124. SEQ ID NO: 20 may optionally contain one additional lysine residue after the terminal valine residue. [103] The one-letter amino acid sequence that corresponds to SEQ ID NO: 17 is shown below:
MDPKGSLSWRILLFLSLAFELSYGQVQLLQPGAELVKPGASVKLACKASGYLFTTY WMHWLKQRPGQGLEWIGEISPTNGRAYYNARFKSEATLTVDKSSNTAYMQLSSLT SEASAVYYCARAYGNYEFAYWGQGTLVTVSVASTKGPSVFPLAPSSKSTSGGTAA LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQT YICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMIS RTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVL HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSL TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQG NVFSCSVMHEALHNHYTQKSLSLSPG*
[104] The one-letter amino acid sequence that corresponds to SEQ ID NO: 18 is shown below:
METDTLLLVWLLLWVPGSTGDWMTQTPLSLPVSLGDQASISCRSRQSLVNSNGN TFLQWYLQKPGQSPKLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGLYF CSQSTHVPPTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGASWCFLNNFYPKDI NVKWKIDGSERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHK TSTSPIVKSFNRNEC*
[105] Positions 21-132 of the above amino acid sequence correspond to SEQ ID NO: 21.
[106] The one-letter amino acid sequence that corresponds to SEQ ID NO: 19 is shown below:
METDTLLLWVLLLVWPGSTGDWMTQTPLSLPVSLGDQASISCRSRQSLVNSNGN TFLQWYLQKPGQSPKLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGLYF CSQSTHVPPTFGGGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREA KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGEC* [107] The one-letter amino acid sequence that corresponds to SEQ ID NO: 22 is shown below:
10 20 30 40 50
ASTKGPSVFP LAPCSRSTSE STAALGCLVK DYFPEPVTVS WNSGALTSGV 60 70 80 90 100
HTFPAVLQSS GLYSLSSWT VPSSNFGTQT YTCNVDHKPS NTKVDKTVER 110 120 130 140 150
KCCVECPPCP APPVAGPSVF LFPPKPKDTL MISRTPEVTC VWDVSHEDP 160 170 180 190
EVQFNWYVDG VEVHNAKTKP REEQFNSTFR WSVLTWHQ
200 210 220 230 240
DWLNGKEYKC KVSNKGLPAP IEKTISKTKG QPREPQVYTL PPSREEMTKN 250 260 270 280 290
QVSLTCLVKG FYPSDISVEW ESNGQPENNY KTTPPMLDSD GSFFLYSKLT 300 310 320
VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK
[108] The one-letter amino acid sequence that corresponds to SEQ ID NO: 23 is SYTMGVS.
[109] The one-letter amino acid sequence that corresponds to SEQ ID NO: 24 is TISSGGGSTYYPDSVKG.
[110] The one-letter amino acid sequence that corresponds to SEQ ID NO: 25 is QGGWLPPFAX, where X may be any naturally occurring amino acid.
[111] The one-letter amino acid sequence that corresponds to SEQ ID NO: 26 is RASKSVSTSSRGYSYMH.
[112] The one-letter amino acid sequence that corresponds to SEQ ID NO: 27 is
LVSNLES. [113] The one-leter amino acid sequence that corresponds to SEQ ID NO: 28 is QHIRELTRS.
[114] The one-leter amino acid sequence that corresponds to SEQ ID NO: 29 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHWVRQAPGQGLEWMGEISPTNGRAYYNQKFQGRVTMTVDKSTNTVYMELSS LRSEDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTA ALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQ TYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMIS RTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVL HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPG.
[115] The DNA sequence that corresponds to SEQ ID NO: 30 is ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTG AAGAAACCTGGCGCCTCCGTGAGGTGTCCTGCAAGGCTTCCGGCTACCTGTTC ACCACCTACTGGATGCACTGGGTGCGACAGGCCCCTGGACAGGGCCTGGAAT GGATGGGCGAGATCTCCCCTACCAACGGCAGAGCCTACTACAACAGAAATTCC AGGGCAGAGTGACCATGACCGTGGACAAGTCCACCAACACCGTGTACATGGAA CTGTCCTCCCTGCGGAGCGAGGACACCGCCGTGTACTACTGCGCTAGAGCCTA CGGCAACTACGATTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCCTC TGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGC ACCAGCGGCGGAACCGCCGCCCTGGGCTGCCTGGGAAGGACTACTTCCCCGA GCCCGTGACCGTGTCCTGGAACAGCGGCGCTCTGACCAGCGGAGTGCACACC TTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTGA CCGTGCCAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACA AGCCCTCCAACACCAAGGTGGACAAGAAGGTGGAGCCTAAGAGCTGCGACAA GACCCACACCTGCCCTCCCTGCCCCGCCCCGAGCTGCTGGGCGGACCCAGCG TGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGCACCCCC GAGGTGACCTGCGTGGTGGTGGACGTGAGCCACGAGGACCCCGAGGTGAGTT CAACTGGTACGTGGACGGCGTGGAGGTGCACAACGCCAAGACCAAGCCTCGG GAGGAGCAGTACAACTCCACCTACCGCGTGGTGAGCGTGCTGACCGTGCTGC ACCAGGACTGGCTGAACGGCAGGAGTACAAGTGCAAGGTGAGCAACAAGGCC CTGCCCGCTCCCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCCCGGG AGCCTCAGGTGTACACCCTGCCCCCCAGCCGCGACGAGCTGACAAGAACCAG GTGAGCCTGACCTGCCTGGTGAAGGGCTTCTACCCCTCCGACATCGCCGTGGA GTGGGAGAGCAACGGCCAGCCTGAGAACAACTACAAGACCACCCCTCCCGTG CTGGACAGCGACGCAGCTTCTTCCTGTACAGCAAGCTGACCGTGGACAAGTCC CGGTGGCAGCAGGGCAACGTGTTCAGCTGCAGCGTGATGCACGAGGCCCTGC ACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCCGGATAGTAA.
[116] The one-letter amino acid sequence that corresponds to SEQ ID NO: 31 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHVWRQAPGQGLEWMGEISPTNGRAYYNAKFQGRVTMTVDKSTNTAYMELSS LRSEDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTA ALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQ TYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMIS RTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVL HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPG.
[117] The DNA sequence that corresponds to SEQ ID NO: 32 is ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTG AAGAAACCTGGCGCCTCCGTGAGGTGTCCTGCAAGGCTTCCGGCTACCTGTTC ACCACCTACTGGATGCACTGGGTGCGACAGGCCCCTGGACAGGGCCTGGAAT GGATGGGCGAGATCTCCCCTACCAACGGCAGAGCCTACTACAACCAAAATTCC AGGGCAGAGTGACCATGACCGTGGACAAGTCCACCAACACCGCTTACATGGAA CTGTCCTCCCTGCGGAGCGAGGACACCGCCGTGTACTACTGCGCTAGAGCCTA CGGCAACTACGATTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCCTC TGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGC ACCAGCGGCGGAACCGCCGCCCTGGGCTGCCTGGGAAGGACTACTTCCCCGA GCCCGTGACCGTGTCCTGGAACAGCGGCGCTCTGACCAGCGGAGTGCACACC TTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTGA CCGTGCCAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACA AGCCCTCCAACACCAAGGTGGACAAGAAGGTGGAGCCTAAGAGCTGCGACAA GACCCACACCTGCCCTCCCTGCCCCGCCCCGAGCTGCTGGGCGGACCCAGCG TGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGCACCCCC GAGGTGACCTGCGTGGTGGTGGACGTGAGCCACGAGGACCCCGAGGTGAGTT CAACTGGTACGTGGACGGCGTGGAGGTGCACAACGCCAAGACCAAGCCTCGG GAGGAGCAGTACAACTCCACCTACCGCGTGGTGAGCGTGCTGACCGTGCTGC ACCAGGACTGGCTGAACGGCAGGAGTACAAGTGCAAGGTGAGCAACAAGGCC CTGCCCGCTCCCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCCCGGG AGCCTCAGGTGTACACCCTGCCCCCCAGCCGCGACGAGCTGACAAGAACCAG GTGAGCCTGACCTGCCTGGTGAAGGGCTTCTACCCCTCCGACATCGCCGTGGA GTGGGAGAGCAACGGCCAGCCTGAGAACAACTACAAGACCACCCCTCCCGTG CTGGACAGCGACGCAGCTTCTTCCTGTACAGCAAGCTGACCGTGGACAAGTCC CGGTGGCAGCAGGGCAACGTGTTCAGCTGCAGCGTGATGCACGAGGCCCTGC ACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCCGGATAGTAA.
[118] The one-leter amino acid sequence that corresponds to SEQ ID NO: 33 is MDPKGSLSWRILLFLSLAFELSYGQVQLVQSGAEVKKPGASVKVSCKASGYLFTTY WMHWVRQAPGQGLEWMGEISPTNGRAYYNAKFQGRVTMTVDKSINTAYMELSRL RSDDTAVYYCARAYGNYFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAA LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQT YICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPPELLGGPSVFLFPPKPKDTLMISR TPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVLH QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELKNQVSLTC LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPG.
[119] The DNA sequence that corresponds to SEQ ID NO: 34 is ATGGACCCCAAGGGCAGCCTGAGCTGGAGAATCCTGCTGTTCCTGAGCCTGGC CTTCGAGCTGAGCTACGGCCAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTG AAGAAACCTGGCGCCTCCGTGAGGTGTCCTGCAAGGCTTCCGGCTACCTGTTC ACCACCTACTGGATGCACTGGGTGCGACAGGCCCCTGGACAGGGCCTGGAAT GGATGGGCGAGATCTCCCCTACCAACGGCAGAGCCTACTACAACCAAAATTCC AGGGCAGAGTGACCATGACCGTGGACAAGTCCATCAACACCGCTTACATGGAA CTGTCCAGACTGCGGAGCGATGACACCGCCGTGTACTACTGCGCTAGAGCCTA CGGCAACTACGATTCGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCCTC TGCTAGCACCAAGGGCCCCAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGC ACCAGCGGCGGAACCGCCGCCCTGGGCTGCCTGGGAAGGACTACTTCCCCGA GCCCGTGACCGTGTCCTGGAACAGCGGCGCTCTGACCAGCGGAGTGCACACC TTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTGA CCGTGCCAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACA AGCCCTCCAACACCAAGGTGGACAAGAAGGTGGAGCCTAAGAGCTGCGACAA GACCCACACCTGCCCTCCCTGCCCCGCCCCGAGCTGCTGGGCGGACCCAGCG TGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGCACCCCC GAGGTGACCTGCGTGGTGGTGGACGTGAGCCACGAGGACCCCGAGGTGAGTT CAACTGGTACGTGGACGGCGTGGAGGTGCACAACGCCAAGACCAAGCCTCGG GAGGAGCAGTACAACTCCACCTACCGCGTGGTGAGCGTGCTGACCGTGCTGC ACCAGGACTGGCTGAACGGCAGGAGTACAAGTGCAAGGTGAGCAACAAGGCC CTGCCCGCTCCCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCCCGGG AGCCTCAGGTGTACACCCTGCCCCCCAGCCGCGACGAGCTGACAAGAACCAG GTGAGCCTGACCTGCCTGGTGAAGGGCTTCTACCCCTCCGACATCGCCGTGGA GTGGGAGAGCAACGGCCAGCCTGAGAACAACTACAAGACCACCCCTCCCGTG CTGGACAGCGACGCAGCTTCTTCCTGTACAGCAAGCTGACCGTGGACAAGTCC CGGTGGCAGCAGGGCAACGTGTTCAGCTGCAGCGTGATGCACGAGGCCCTGC ACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCCGGATAGTAA.
[120] The one-leter amino acid sequence that corresponds to SEQ ID NO: 35 is METDTLLLWVLLLWVPGSTGDWMTQSPLSLPVTLGQPASISCRSSQSLVNSNGNT FLQWYQQRPGQSPRLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY CSQSTHVPPTFGGGTVEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAK VQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG LSSPVTKSFNRGEC.
[121] The DNA sequence that corresponds to SEQ ID NO: 36 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCCTGCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCTCCCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCAGCAGCGGCCTGGCCAGAGCCCCAGAC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCG GCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CGGAGGCACCAAGTGGAAATCAAGCGGACCGTGGCCGCCCCCAGCGTGTTCA TCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGTG CCTGCTGAACAACTTCTACCCCCGCGAGGCCAAGGGCAGTGGAAGGTGGACA ACGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGACTCCAA GGACAGCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTAC GAGAAGACAAGGTGTACGCCTGCGAGGTGACCCACCAGGGACTGTCTAGCCC CGTGACCAAGAGCTTCAACCGGGGCGAGTGCTAA.
[122] The one-letter amino acid sequence that corresponds to SEQ ID NO: 37 is METDTLLLWVLLLWVPGSTGDWMTQSPLSLPVTLGQPASISCRSRQSLVNSNGN TFLQWYQQRPGQSPRLLIYKVSLRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVY YCSQSTHVPPTFGGGTVEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREA KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGEC.
[123] The DNA sequence that corresponds to SEQ ID NO: 38 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCCTGCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCAGGCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCAGCAGCGGCCTGGCCAGAGCCCCAGAC TGCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCG GCTCTGGCTCTGGCACCGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CGGAGGCACCAAGTGGAAATCAAGCGGACCGTGGCCGCCCCCAGCGTGTTCA TCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGTG CCTGCTGAACAACTTCTACCCCCGCGAGGCCAAGGGCAGTGGAAGGTGGACA ACGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGACTCCAA GGACAGCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTAC
GAGAAGACAAGGTGTACGCCTGCGAGGTGACCCACCAGGGACTGTCTAGCCC
CGTGACCAAGAGCTTCAACCGGGGCGAGTGCTAA.
[124] The one-leter amino acid sequence that corresponds to SEQ ID NO: 39 is METDTLLLWVLLLWVPGSTGDWMTQSPLSSPVTLGQPASISCRSSQSLVNSNGN TFLQWYHQRPGQPPRLLIYKVSLRFSGVPDRFSGSGAGKDFTLKISRVEAEDVGVY YCSQSTHVPPTFGQGTLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREA KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGEC.
[125] The DNA sequence that corresponds to SEQ ID NO: 40 is ATGGAGACCGACACCCTGCTGCTCTGGGTGCTGCTGCTCTGGGTGCCCGGCT CCACCGGAGACGTCGTGATGACCCAGTCCCCTCTGTCCAGTCCTGTGACCCTG GGACAGCCTGCCTCCATCTCCTCAGATCCTCCCAGTCCCTCGTGAACTCCAAC GGCAACACCTTCCTGCAGTGGTATCACCAGCGGCCTGGCCAGCCTCCCAGACT GCTGATCTACAAGGTGTCCCTGCGGTTCTCCGGCGTGCCCGACGATTTTCCGG CTCTGGCGCTGGCAAGGACTTCACCCTGAAGATCTCCCGGGTGGAAGCCGAG GACGTGGGCGTGTACTACTGCTCCCAGAGCACCCACGTGCCCCCTACATTTGG CCAGGGCACCAACTGGAAATCAAGCGGACCGTGGCCGCCCCCAGCGTGTTCA TCTTCCCTCCCAGCGACGAGCAGCTGAAGTCTGGCACCGCCAGCGTGGTGTG CCTGCTGAACAACTTCTACCCCCGCGAGGCCAAGGGCAGTGGAAGGTGGACA ACGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGACTCCAA GGACAGCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTAC GAGAAGACAAGGTGTACGCCTGCGAGGTGACCCACCAGGGACTGTCTAGCCC CGTGACCAAGAGCTTCAACCGGGGCGAGTGCTAA.
[126] EXAMPLES
[127] Example 1 : In vivo study of the administration of anti-glycation end-product antibody
[128] To examine the effects of an anti-glycation end-product antibody, the antibody was administered to the aged CD1(ICR) mouse (Charles River Laboratories), twice daily by intravenous injection, once a week, for three weeks (Days 1 , 8 and 15), followed by a 10 week treatment-free period. The test antibody was a commercially available mouse anti-glycation end-product antibody raised against carboxymethyl lysine conjugated with keyhole limpet hemocyanin, the carboxymethyl lysine MAb (Clone 318003) available from R&D Systems, Inc. (Minneapolis, MN; catalog no. MAB3247). A control reference of physiological saline was used in the control animals.
[129] Mice referred to as “young” were 8 weeks old, while mice referred to as “old” were 88 weeks (±2 days) old. No adverse events were noted from the administration of the antibody. The different groups of animals used in the study are shown in Table 1.
[130] Table 1 : The different groups of animals used in the study
Figure imgf000045_0001
- = Not Applicable, Pre = Subset of animals euthanized prior to treatment start for collection of adipose tissue.
[131] p-jgiNK4a mRNA, a marker for senescent cells, was quantified in adipose tissue of the groups by Real Time-qPCR. The results are shown in Table 2. In the table AACt = ACt mean control Group (2) - ACt mean experimental Group (1 or 3 or 5); Fold Expression= 2 -AACt.
[132] Table 2: pi6INK4a mRNA quantified in adipose tissue
Figure imgf000045_0002
Figure imgf000046_0001
[133] The table above indicates that untreated old mice (Control Group 2) express 2.55-fold more p16lnk4a mRNA than the untreated young mice (Control Group 1), as expected. This was observed when comparing Group 2 untreated old mice euthanized at end of recovery Day 85 to Group 1 untreated young mice euthanized at end of treatment Day 22. When results from Group 2 untreated old mice were compared to results from Group 3 treated old mice euthanized Day 85, it was observed that p16lnk4a mRNA was 1.23-fold higher in Group 2 than in Group 3. Therefore, the level of p16lnk4a mRNA expression was lower when the old mice were treated with 2.5 pg/gram/BID/week of antibody.
[134] When results from Group 2 (Control) untreated old mice were compared to results from Group 5 (5 pg/gram) treated old mice euthanized Day 22, it was observed that p16lnk4a mRNA was 3.03-fold higher in Group 2 (controls) than in Group 5 (5 pg/gram). This comparison indicated that the Group 5 animals had lower levels of p16lnk4a mRNA expression when they were treated with 5.0 pg/gram/BID/week, providing p16lnk4a mRNA expression levels comparable to that of the young untreated mice (i.e. Group 1). Unlike Group 3 (2.5 pg/gram) mice that were euthanized at end of recovery Day 85, Group 5 mice were euthanized at end of treatment Day 22.
[135] These results indicate the antibody administration resulted in the killing of senescent cells.
[136] The mass of the gastrocnemius muscle was also measured, to determine the effect of antibody administration on sarcopenia. The results are provided in Table 3. The results indicate that administration of the antibody increased muscle mass as compared to controls, but only at the higher dosage of 5.0 pg/gm/BID/ week. [137] Table 3: Effect of antibody administration on mass of the gastrocnemius muscle
Weight relative to body
Summary Absolute weight of mass of Gastrocnemius Group Information Gastrocnemius Muscle Muscle
1 Mean 0.3291 1.1037
SD 0.0412 0.1473
N 20 20
2 Mean 0.3304 0.7671
SD 0.0371 0.1246
N 20 20
3 Mean 0.3410 0.7706
SD 0.0439 0.0971
N 19 19
5 Mean 0.4074 0.9480
SD 0.0508 0.2049
N 9 9
[138] These results demonstrate that administration of antibodies that bind to AGEs of a cell resulted in a reduction of cells expressing p16lnk4a, a biomarker of senescence. The data show that reducing senescent cells leads directly to an increase in muscle mass in aged mice. These results indicate that the loss of muscle mass, a classic sign of sarcopenia, can be treated by administration of antibodies that bind to AGEs of a cell. The results suggest that administration of the antibodies would be effective in treating osteoarthritis by removing senescent chondrocytes.
[139] Example 2: Affinity and kinetics of test antibody
[140] The affinity and kinetics of the test antibody used in Example 1 were analyzed using Na,Na-bis(carboxymethyl)-L-lysine trifluoroacetate salt (Sigma-Aldrich, St. Louis, MO) as a model substrate for an AGE-modified protein of a cell. Label-free interaction analysis was carried out on a BIACORE™ T200 (GE Healthcare, Pittsburgh, PA), using a Series S sensor chip CM5 (GE Healthcare, Pittsburgh, PA), with Fc1 set as blank, and Fc2 immodilized with the test antibody (molecular weigh of 150,000 Da). The running buffer was a HBS-EP buffer (10 mM HEPES, 150 mM NaCI, 3 mM EDTA and 0.05% P-20, pH of 7.4), at a temperature of 25 °C. Software was BIACORE™ T200 evaluation software, version 2.0. A double reference (Fc2-1 and only buffer injection), was used in the analysis, and the data was fitted to a Langmuir 1:1 binding model.
[141] Table 4: Experimental set-up of affinity and kinetics analysis
Figure imgf000048_0001
[142] A graph of the response versus time is illustrated in FIG. 1. The following values were determined from the analysis: ka (1/Ms) = 1.857 x 103; kd (1/s) = 6.781 x 10‘3; KD (M) = 3.651 x 10'6; Rmax (RU) = 19.52; and Chi2 = 0.114. Because the Chi2 value of the fitting is less than 10% of Rmax, the fit is reliable.
[143] Example 3: Construction and production of murine anti-AGE lgG2b antibody and chimeric anti-AGE lgG1 antibody
[144] Murine and chimeric human anti-AGE antibodies were prepared. The DNA sequence of murine anti-AGE antibody lgG2b heavy chain is shown in SEQ ID NO: 12. The DNA sequence of chimeric human anti-AGE antibody lgG1 heavy chain is shown in SEQ ID NO: 13. The DNA sequence of murine anti-AGE antibody kappa light chain is shown in SEQ ID NO: 14. The DNA sequence of chimeric human anti- AGE antibody kappa light chain is shown in SEQ ID NO: 15. The gene sequences were synthesized and cloned into high expression mammalian vectors. The sequences were codon optimized. Completed constructs were sequence confirmed before proceeding to transfection.
[145] HEK293 cells were seeded in a shake flask one day before transfection, and were grown using serum-free chemically defined media. The DNA expression constructs were transiently transfected into 0.03 liters of suspension HEK293 cells. After 20 hours, cells were sampled to obtain the viabilities and viable cell counts, and titers were measured (Octet QKe, ForteBio). Additional readings were taken throughout the transient transfection production runs. The cultures were harvested on day 5, and an additional sample for each was measured for cell density, viability and titer.
[146] The conditioned media for murine and chimeric anti-AGE antibodies were harvested and clarified from the transient transfection production runs by centrifugation and filtration. The supernatants were run over a Protein A column and eluted with a low pH buffer. Filtration using a 0.2 pm membrane filter was performed before aliquoting. After purification and filtration, the protein concentrations were calculated from the OD280 and the extinction coefficient. A summary of yields and aliquots is shown in Table 5:
[147] Table 5: Yields and aliquots
Figure imgf000049_0001
[148] Antibody purity was evaluated by capillary electrophoresis sodium-dodecyl sulfate (CE-SDS) analysis using LabChip® GXII, (PerkinElmer).
[149] Example 4: Binding of murine (parental) and chimeric anti-AGE antibodies
[150] The binding of the murine (parental) and chimeric anti-AGE antibodies described in Example 3 was investigated by a direct binding ELISA. An anti- carboxymethyl lysine (CML) antibody (R&D Systems, MAB3247) was used as a control. CML was conjugated to KLH (CML-KLH) and both CML and CML-KLH were coated overnight onto an ELISA plate. HRP-goat anti-mouse Fc was used to detect the control and murine (parental) anti-AGE antibodies. HRP-goat anti-human Fc was used to detect the chimeric anti-AGE antibody.
[161] The antigens were diluted to 1 pg/mL in 1x phosphate buffer at pH 6.5. A 96- well microtiter ELISA plate was coated with 100 pL/well of the diluted antigen and let sit at 4°C overnight. The plate was blocked with 1x PBS, 2.5% BSA and allowed to sit for 1-2 hours the next morning at room temperature. The antibody samples were prepared in serial dilutions with 1x PBS, 1% BSA with the starting concentration of 50 pg/mL. Secondary antibodies were diluted 1 :5,000. 100 pL of the antibody dilutions was applied to each well. The plate was incubated at room temperature for 0.5-1 hour on a microplate shaker. The plate was washed 3 times with 1x PBS. 100 pL/well diluted HRP-conjugated goat anti-human Fc secondary antibody was applied to the wells. The plate was incubated for 1 hour on a microplate shaker. The plate was then washed 3 times with 1x PBS. 100 pL HRP substrate TMB was added to each well to develop the plate. After 3-5 minutes elapsed, the reaction was terminated by adding 100 pL of 1 N HCL A second direct binding ELISA was performed with only CML coating. The absorbance at OD450 was read using a microplate reader.
[152] The OD450 absorbance raw data for the CML and CML-KLH ELISA is shown in the plate map below. 48 of the 96 wells in the well plate were used. Blank wells in the plate map indicate unused wells. [153] Plate map of CML and CML-KLH ELISA:
Cone.
(ug/mL) 1 2 3 4 5 6 7
Figure imgf000051_0001
R&D Parental Chimeric R&D Parental Chimeric
Positive Anti- Anti- Positive Anti- Anti-
Control AGE AGE Control AGE AGE
CML-KLH Coat CML Coat
[154] The OD450 absorbance raw data for the CML-only ELISA is shown in the plate map below. 24 of the 96 wells in the well plate were used. Blank wells in the plate map indicate unused wells.
[155] Plate map of CML-only ELISA:
Cone.
(ug/mL) 1 2 3 4 5 6 7
Figure imgf000051_0002
R&D Parental Chimeric
Positive Anti- Anti¬
Control AGE AGE [156] The control and chimeric anti-AGE antibodies showed binding to both CML and CML-KLH. The murine (parental) anti-AGE antibody showed very weak to no binding to either CML or CML-KLH. Data from repeated ELISA confirms binding of the control and chimeric anti-AGE to CML. All buffer control showed negative signal.
[157] Example 5: Humanized antibodies
[158] Humanized antibodies were designed by creating multiple hybrid sequences that fuse select parts of the parental (mouse) antibody sequence with the human framework sequences. Acceptor frameworks were identified based on the overall sequence identity across the framework, matching interface position, similarly classed CDR canonical positions, and presence of N-glycosylation sites that would have to be removed. Three humanized light chains and three humanized heavy chains were designed based on two different heavy and light chain human acceptor frameworks. The amino acid sequences of the heavy chains are shown in SEQ ID NO: 29, 31 and 33, which are encoded by the DNA sequences shown in SEQ ID NO: 30, 32 and 34, respectively. The amino acid sequences of the light chains are shown in SEQ ID NO: 35, 37 and 39, which are encoded by the DNA sequences shown in SEQ ID NO: 36, 38 and 40, respectively. The humanized sequences were methodically analyzed by eye and computer modeling to isolate the sequences that would most likely retain antigen binding. The goal was to maximize the amount of human sequence in the final humanized antibodies while retaining the original antibody specificity. The light and heavy humanized chains could be combined to create nine variant fully humanized antibodies.
[159] The three heavy chains and three light chains were analyzed to determine their humanness. Antibody humanness scores were calculated according to the method described in Gao, S. H., et al., “Monoclonal antibody humanness score and its applications”, BMC Biotechnology, 13:55 (July 5, 2013). The humanness score represents how human-like an antibody variable region sequence looks. For heavy chains a score of 79 or above is indicative of looking human-like; for light chains a score of 86 or above is indicative of looking human-like. The humanness of the three heavy chains, three light chains, a parental (mouse) heavy chain and a parental (mouse) light chain are shown below in Table 6:
[160] Table 6: Antibody humanness
Figure imgf000053_0001
[161] Full-length antibody genes were constructed by first synthesizing the variable region sequences. The sequences were optimized for expression in mammalian cells. These variable region sequences were then cloned into expression vectors that already contain human Fc domains; for the heavy chain, the lgG1 was used.
[162] Small scale production of humanized antibodies was carried out by transfecting plasmids for the heavy and light chains into suspension HEK293 cells using chemically defined media in the absence of serum. Whole antibodies in the conditioned media were purified using MabSelect SuRe Protein A medium (GE Healthcare). [163] Nine humanized antibodies were produced from each combination of the three heavy chains having the amino acid sequences shown in SEQ ID NO: 29, 31 and 33 and three light chains having the amino acid sequences shown in SEQ ID NO: 35, 37 and 39. A comparative chimeric parental antibody was also prepared. The antibodies and their respective titers are shown below in Table 7:
[164] Table 7: Antibody titers
Figure imgf000054_0001
[165] The binding of the humanized antibodies may be evaluated, for example, by dose-dependent binding ELISA or cell-based binding assay.
[166] Example 6 (Prophetic): An AGE-RNAse containing vaccine in a human subject. [167] AGE-RNAse is prepared by incubating RNAse in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-RNAse solution is dialyzed and the protein content is measured. Aluminum hydroxide or aluminum phosphate, as an adjuvant, is added to 100 pg of the AGE-RNAse. Formaldehyde or formalin is added as a preservative to the preparation. Ascorbic acid is added as an antioxidant. The vaccine also includes phosphate buffer to adjust the pH and glycine as a protein stabilizer. The composition is injected intravenously into a subject with osteoarthritis.
[168] Example 7 (Prophetic): Injection regimen for an AGE-RNAse containing vaccine in a human subject
[169] The same vaccine as described in Example 6 is injected intra-articularly into a subject with osteoarthritis. The titer of antibodies to AGE-RNAse is determined by ELISA after two weeks. Additional injections are performed after three weeks and six weeks, respectively. Further titer determination is performed two weeks after each injection.
[170] Example 8 (Prophetic): An AGE-hemoglobin containing vaccine in a human subject.
[171] AGE-hemoglobin is prepared by incubating human hemoglobin in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-hemoglobin solution is dialyzed and the protein content is measured. All vaccine components are the same as in Example 6, except AGE-hemoglobin is substituted for AGE-RNAse. Administration is carried out as in Example 6, or as in Example 7.
[172] Example 9 (Prophetic): An AGE-human serum albumin containing vaccine in a human subject.
[173] AGE-human serum albumin is prepared by incubating human serum albumin in a phosphate buffer solution containing 0.1-3 M glucose, glucose-6-phosphate, fructose or ribose for 10-100 days. The AGE-human serum albumin solution is dialyzed and the protein content is measured. All vaccine components are the same as in Example 6, except AGE-human serum albumin is substituted for AGE-RNAse. Administration is carried out as in Example 6, or as in Example 7.
[174] Example 10: Carboxymethyllysine-modified protein vaccine for a human subject (prophetic)
[175] A vaccine is prepared by combining a carboxymethyllysine-modified protein as an AGE antigen, aluminum hydroxide as an adjuvant, formaldehyde as a preservative, ascorbic acid as an antioxidant, a phosphate buffer to adjust the pH of the vaccine and glycine as a protein stabilizer. The vaccine is injected subcutaneously into a subject with osteoarthritis.
[176] Example 11 : Carboxyethyllysine-modified peptide vaccine for a human subject (prophetic)
[177] A vaccine is prepared by combining a carboxyethyllysine-modified peptide conjugated to KLH as an AGE antigen, aluminum hydroxide as an adjuvant, formaldehyde as a preservative, ascorbic acid as an antioxidant, a phosphate buffer to adjust the pH of the vaccine and glycine as a protein stabilizer. The vaccine is injected subcutaneously into a subject with osteoarthritis.
[178] Example 12: In vivo study of the administration of a carboxymethyl lysine monoclonal antibody
[179] The effect of a carboxymethyl lysine antibody on tumor growth, metastatic potential and cachexia was investigated. In vivo studies were carried out in mice using a murine breast cancer tumor model. Female BALB/c mice (BALB/cAnNCrl, Charles River) were eleven weeks old on Day 1 of the study.
[180] 4T1 murine breast tumor cells (ATCC CRL-2539) were cultured in RPMI 1640 medium containing 10% fetal bovine serum, 2 mM glutamine, 25 pg/mL gentamicin, 100 units/mL penicillin G Na and 100 pg/mL streptomycin sulfate. Tumor cells were maintained in tissue culture flasks in a humidified incubator at 37 °C in an atmosphere of 5% CO2 and 95% air. [181] The cultured breast cancer cells were then implanted in the mice. 4T1 cells were harvested during log phase growth and re-suspended in phosphate buffered saline (PBS) at a concentration of 1 x 106 cells/mL on the day of implant. Tumors were initiated by subcutaneously implanting 1 x 1054T1 cells (0.1 mL suspension) into the right flank of each test animal. Tumors were monitored as their volumes approached a target range of 80-120 mm3. Tumor volume was determined using the formula: tumor volume = (tumor width)2(tumor length)/2. Tumor weight was approximated using the assumption that 1 mm3 of tumor volume has a weight of 1 mg. Thirteen days after implantation, designated as Day 1 of the study, mice were sorted into four groups (n=15/group) with individual tumor volumes ranging from 108 to 126 mm3 and a group mean tumor volume of 112 mm3. The four treatment groups are shown in Table 8 below:
[182] Table 8: Treatment groups
Figure imgf000057_0001
[183] An anti-carboxymethyl lysine monoclonal antibody was used as a therapeutic agent 250 mg of carboxymethyl lysine monoclonal antibody was obtained from R&D Systems (Minneapolis, MN). Dosing solutions of the carboxymethyl lysine monoclonal antibody were prepared at 1 and 0.5 mg/mL in a vehicle (PBS) to provide the active dosages of 10 and 5 pg/g, respectively, in a dosing volume of 10 mL/kg. Dosing solutions were stored at 4 °C protected from light.
[184] All treatments were administered intravenously (i.v.) twice daily for 21 days, except on Day 1 of the study where the mice were administered one dose. On Day 19 of the study, i.v. dosing was changed to intraperitoneal (i.p.) dosing for those animals that could not be dosed i.v. due to tail vein degradation. The dosing volume was 0.200 mL per 20 grams of body weight (10 mL/kg), and was scaled to the body weight of each individual animal.
[185] The study continued for 23 days. Tumors were measured using calipers twice per week. Animals were weighed daily on Days 1-5, then twice per week until the completion of the study. Mice were also observed for any side effects. Acceptable toxicity was defined as a group mean body weight loss of less than 20% during the study and not more than 10% treatment-related deaths. Treatment efficacy was determined using data from the final day of the study (Day 23).
[186] The ability of the anti-carboxymethyl lysine antibody to inhibit tumor growth was determined by comparing the median tumor volume (MTV) for Groups 1-3. Tumor volume was measured as described above. Percent tumor growth inhibition (%TGI) was defined as the difference between the MTV of the control group (Group 1) and the MTV of the drug-treated group, expressed as a percentage of the MTV of the control group. %TGI may be calculated according to the formula: %TGI = (1- MTVtreated/MTVcontrol) X 100.
[187] The ability of the anti-carboxymethyl lysine antibody to inhibit cancer metastasis was determined by comparing lung cancer foci for Groups 1-3. Percent inhibition (%lnhibition) was defined as the difference between the mean count of metastatic foci of the control group and the mean count of metastatic foci of a drug- treated group, expressed as a percentage of the mean count of metastatic foci of the control group. %lnhibition may be calculated according to the following formula: %lnhibition = (1-Mean Count of Focitreated/Mean Count of Focicontroi) x 100. [188] The ability of the anti-carboxymethyl lysine antibody to inhibit cachexia was determined by comparing the weights of the lungs and gastrocnemius muscles for Groups 1-3. Tissue weights were also normalized to 100 g body weight.
[189] Treatment efficacy was also evaluated by the incidence and magnitude of regression responses observed during the study. Treatment may cause partial regression (PR) or complete regression (CR) of the tumor in an animal. In a PR response, the tumor volume was 50% or less of its Day 1 volume for three consecutive measurements during the course of the study, and equal to or greater than 13.5 mm3 for one or more of these three measurements. In a CR response, the tumor volume was less than 13.5 mm3 for three consecutive measurements during the course of the study.
[190] Statistical analysis was carried out using Prism (GraphPad) for Windows 6.07. Statistical analyses of the differences between Day 23 mean tumor volumes (MTVs) of two groups were accomplished using the Mann-Whitney U test. Comparisons of metastatic foci were assessed by ANOVA-Dunnett. Normalized tissue weights were compared by ANOVA. Two-tailed statistical analyses were conducted at significance level P = 0.05. Results were classified as statistically significant or not statistically significant.
[191] The results of the study are shown below in Table 9:
[192] Table 9: Results
Figure imgf000059_0001
Figure imgf000060_0001
[193] All treatment regimens were acceptably tolerated with no treatment-related deaths. The only animal deaths were non-treatment-related deaths due to metastasis. The %TGI was not statistically significant (P > 0.05, Mann-Whitney) for the 5 pg/g (Group 2) or 10 pg/g treatment group (Group 3). The %lnhibition was not statistically significant (P > 0.05, ANOVA-Dunnett) for the 5 pg/g treatment group. The %lnhibition was statistically significant (P < 0.01 , ANOVA-Dunnett) for the 10 pg/g treatment group. Although the statistical significance of the cachexia inhibition could have been greater (P > 0.05, ANOVA), the results indicate that administration of an anti-carboxymethyl lysine monoclonal antibody is able to improve cancer cachexia. This data provides additional evidence that in vivo administration of anti- AGE antibodies can provide therapeutic benefits safely and effectively.
[194] Example 13: In vivo osteoarthritis study (prophetic)
[195] In vivo studies are carried out in mice to study the effect of treatment with anti- AGE antibodies and vaccination with AGE-KLH on osteoarthritis. Male C57/BL6 mice are 8-10 weeks old on Day 1 of the study. The mice are separated into five treatment groups: (1) control; (2) vehicle only administered intravenously; (3) anti- AGE antibody at 10 pg/g dose administered intravenously; (4) anti-AGE antibody at 10 pg/g dose administered intra-articularly; and (5) 10 pg AGE-KLH administered as a vaccine intraperitoneally.
[196] Osteoarthritis is induced in Groups 2-5 by medial meniscal destabilization of the right hind paw knee. Group 1 is a non-arthritic control is sham operated on by exposure of the left hind paw medial meniscotibial ligament with no transection. All animals are anesthetized with isoflurane, shaved and prepared for surgery. In Groups 2-5 destabilization is achieved by the complete rupture of the medial meniscus ligament on the left joint. The knee capsule is closed with absorbable suture material. Skin is closed with a subcutaneous suture of absorbable suture and surgical glue. Buprenorphine (0.3mg/ml) is provided at a dose level of 0.1mg/kg presurgery and 8-12 hours post first injection.
[197] Dosing begins one week after the surgery. For Groups 2-5, the dosing volume is 0.200 mL per 20 grams of body weight (10 mL/kg), and is scaled to the body weight of each individual animal. Group 2 receives phosphate-buffered saline (PBS) delivered intravenously. Group 3 receives 10pg/g of an anti-AGE antibody twice daily for 21 days delivered intravenously. Group 4 receives 10pg/g of an anti- AGE antibody twice daily for 21 days delivered intra-articularly into the knee that was operated on. Group 5 receives 10pg of AGE-KLH in Freunds complete adjuvant intraperitoneally one week prior to inducing osteoarthritis, followed by a 10pg/g booster injection of the vaccine four weeks after surgery.
[198] All Groups are monitored daily for morbidity/mortality and are evaluated daily with attention to effects on locomotion and altered gait. Osteoarthritis pain is measured in all groups by dynamic weight bearing (DMB) testing, a standard test for osteoarthritis in rodents.
[199] The animals in Groups 1 and 5 are sacrificed at week 16. For Group 5, the blood is collected for an antibody titer assay, such as the THERMOFISHER® EASY- TITER® Mouse IgG Assay, to determine the titer of antibody in the mice specific for anti-AGE antibodies. An equal number of animals in Groups 2-4 are sacrificed at weeks 4, 8 and 16. Half of the mice in each sacrificed group are analyzed for histology and half are analyzed for p16INK4a qRT PCR. p16INK4a is measured in articular cartilage (chondrocytes) of the animals sacrificed. The p16INK4a qRT PCR is preserved for qRT PCR analysis.
[200] Osteoarthritis severity is also measured by evaluating samples of the knee joints. Sample of the right and left whole knee joints from all mice are collected and fixed in 10% NBF, then decalcified and embedded in paraffin wax. Three non- consecutive coronal sections are taken for the right knee joint and another three non- consecutive coronal sections are taken for the left knee joint for each staining, providing 6 slides per animal for each stain for a total of 12 slides per animal. The sections are scored for disease severity (cartilage/bone with osteophytes and synovial membrane) by a board certified veterinary pathologist using a semi- quantitative grading system. Scores are reported with statistical analysis.
[201] The anti-AGE antibody will specifically bind to senescent chondrocytes and allow the immune system to destroy those cells. Similarly, vaccination with an AGE- KLH antigen will allow the murine immune system to target and remove senescent chondrocytes. Killing and removing senescent chondrocytes will prevent the development of osteoarthritis.
[202] Example 14: Anti-AGE antibodies bind to senescent chondrocytes in vitro
[203] Senescent chondrocytes were obtained from osteoarthritic joints. Anti-AGE antibodies bound to the senescent chondrocytes in vitro. These results suggest that anti-AGE antibodies could be administered therapeutically in vivo to treat osteoarthritis by specifically targeting senescent chondrocytes.
[204] Example 15: Summary of Jeon et al., 2017, which is incorporated herein by reference
[205] To determine whether senescent cells contribute to osteoarthritis pathogenesis, the development of senescent cells was studied following articular joint injury. Post-traumatic osteoarthritis was induced by anterior cruciate ligament transection (ACLT) in p16-3MR mice. P16-3MR mice harbor p16INK4a promoter, which drives the expression of a fusion protein containing synthetic Renilla luciferase and monomeric red fluorescent protein domains, for non-invasive monitoring. Luminescence increased in the articular joint region in the p16-3MR mice after the ACLT surgery.
[206] To determine whether selective removal of senescent cells that develop after the ACLT would reduce or reverse the progression of osteoarthritis, the senescent cells were selectively removed. Selective killing of senescent cells was achieved by administering ganciclovir to the p16-3MR mice, which is effective due to the fact that the p16-3MR transgene includes a truncated form of herpes simplex virus 1 thymidine kinase (HSV-TK). The clearance resulted in a reduction in p16INK4a expression. Clearance of senescent cells inhibited articular cartilage erosion.
[207] REFERENCES
[208] 1. International Application Pub. No. WO 2009/143411 to Gruber (26 Nov. 2009).
[209] 2. U.S. Patent No. 5,702,704 to Bucala (issued December 30, 1997).
[210] 3. U.S. Patent No. 6,380,165 to Al-Abed et al. (issued April 30, 2002).
[211] 4. U.S. Patent No. 6,387,373 to Wright et al. (issued May 14, 2002).
[212] 5. U.S. Patent No. 4,217,344 to Vanlerberghe et al. (issued August 12,
1980).
[213] 6. U.S. Patent No. 4,917,951 to Wallach (issued April 17, 1990).
[214] 7. U.S. Patent No. 4,911 ,928 to Wallach (issued March 27, 1990).
[215] 8. U.S. Patent Application Publication Pub. No. US 2010/226932 to Smith etal. (September 9, 2010).
[216] 9. Baker, D.J. et al., “Clearance of p16lnk4a-positive senescent cells delays ageing-associated disorders”, Nature, Vol. 479, pp. 232-236, (2011).
[217] 10. Ando, K. et al., “Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation”, Biochem. Biophys. Res. Commun., Vol. 258, 123, 125 (1999).
[218] 11. Lindsey, J. B. et al., “Receptor For Advanced Glycation End-Products (RAGE) and soluble RAGE (sRAGE): Cardiovascular Implications", Diabetes Vascular Disease Research, Vol. 6(1), 7-14, (2009).
[219] 12. Bierhaus, A., “AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept”, Cardiovasc. Res., Vol. 37(3), 586-600 (1998). [220] 13. Ahmed, E. K. et al., “Protein Modification and Replicative Senescence of WI-38 Human Embryonic Fibroblasts", Aging Cells, Vol. 9, 252, 260 (2010).
[221] 14. Vlassara, H. et al., “Advanced Glycosylation Endproducts on Erythrocyte Cell Surface Induce Receptor-Mediated Phagocytosis by Macrophages”, J. Exp. Med., Vol. 166, 539, 545 (1987).
[222] 15. Vlassara, H. et al., “High-affinity-receptor-mediated Uptake and Degradation of Glucose-modified Proteins: A Potential Mechanism for the Removal of Senescent Macromolecules”, Proc. Natl. Acad. Sci. USA, Vol. 82, 5588, 5591 (1985).
[223] 16. Roll, P. et al., “Anti-CD20 Therapy in Patients with Rheumatoid Arthritis”, Arthritis & Rheumatism, Vol. 58, No. 6, 1566-1575 (2008).
[224] 17. Kajstura, J. et al., “Myocite Turnover in the Aging Human Heart”, Circ. Res., Vol. 107(11), 1374-86, (2010).
[225] 18. de Groot, K. et al., “Vascular Endothelial Damage and Repair in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis”, Arthritis and Rheumatism, Vol. 56(11), 3847, 3847 (2007).
[226] 19. Manesso, E. et al., “Dynamics of P-Cell Turnover: Evidence for p-Cell Turnover and Regeneration from Sources of p-Cells other than p-cell Replication in the HIP Rat”, Am. J. Physiol. Endocrinol. Metab., Vol. 297, E323, E324 (2009).
[227] 20. Kirstein, M. et al., “Receptor-specific Induction of Insulin-like Growth Factor I in Human Monocytes by Advanced Glycosylation End Product-modified Proteins”, J. Clin. Invest., Vol. 90, 439, 439-440 (1992).
[228] 21. Murphy, J. F., “Trends in cancer immunotherapy”, Clinical Medical Insights: Oncology, Vol. 14(4), 67-80 (2010).
[229] 22. Flint, S. J. etal., “Principles of Virology”, ASM Press (2000). [230] 23. Buskas, T. et al., “Immunotherapy for Cancer: Synthetic Carbohydrate- based Vaccines”, Chem. Commun., Vol. 28(36), 5335-349 (2009).
[231] 24. Beier, K. C. et al., “Master Switches of T-cell Differentiation”, Eur. Respir. J., Vol. 29, 804-12 (2007).
[232] 25. Schmidlin H. et al., “New Insights in the Regulation of Human B Cell Differentiation”, Trends Immunol., Vol. 30(6), 277-85 (2009).
[233] 26. Vogel, F. R. et al., “A compendium of vaccine adjuvants and excipients”, Pharmaceutical Biotechnology, Vol. 6, pp. 141-228 (1995).
[234] 27. Coler, R. N. et al., “Development and Characterization of Synthetic Glucopyranosyl Lipid Adjuvant System as a Vaccine Adjuvant”, PLoS ONE, Vol. 6(1): e16333 (2011).
[235] 28. Cheadle, E. J. et al., “Bugs as Drugs for Cancer”, Immunology, Vol. 107, 10-19 (2002).
[236] 29. Jiang, Z-H. et al. “Synthetic vaccines: the role of adjuvants in immune targeting”, Current Medicinal Chemistry, Vol. 10(15), pp. 1423-39 (2003).
[2371 30. Virella, G. et al., “Autoimmune Response to Advanced Glycosylation
End-Products of Human LDL”, Journal of Lipid Research, Vol. 44, 487-493 (2003).
[238] 31. Ameli, S. et al. , “Effect of Immunization With Homologous LDL and Oxidized LDL on Early Atherosclerosis in Hypercholesterolemic Rabbits”, Arteriosclerosis, Thrombosis, and Vascular Biology, Vol. 16, 1074 (1996).
[239] 32. “Vaccine Excipient & Media Summary", available online at www.cdc.gov/vaccines/pubs/pinkbook/downloads/appendices/B/excipient-table-2.pdf (The Pink Book, Epidemiology and Prevention of Vaccine-Preventable Diseases, 12th Ed. Second Printing, September 2013).
[240] 33. “Sarcopenia”, available online at en.wikipedia.org/wiki/Sarcopenia (November 14, 2014). [241] 34. “What is sarcopenia?”, available online at www.iofbonehealth.org/what- sarcopenia (2014).
[242] 35. Blahd, W., “Sarcopenia with aging”, available online at www.webmd.com/healthy-aging/sarcopenia-with-aging (August 3, 2014).
[243] 36. “Keyhole limpet hemocyanin”, available online at en.wikipedia.org/wiki/Keyhole_limpet_hemocyanin (April 18, 2014).
[244] 37. “CML-BSA Product Data Sheet’, available online at www.cellbiolabs.com/sites/default/files/STA-314-cml-bsa.pdf (2010).
[245] 38. “CML (N-epsilon-(Carboxymethyl)Lysine) Assays and Reagents”, available online atwww.cellbiolabs.com/cml-assays (Accessed on December 15, 2014).
[246] 39. Cruz-Jentoft, A. J. et al., “Sarcopenia: European consensus on definition and diagnosis”, Age and Ageing, Vol. 39, pp. 412-423 (April 13, 2010).
[247] 40. Rolland, Y. et al., “Sarcopenia: its assessment, etiology, pathogenesis, consequences and future perspectives", J. Nutr. Health Aging, Vol. 12(7), pp. 433- 450 (2008).
[248] 41. Mera, K. et al., “An autoantibody against N£-(carboxyethyl)lysine (CEL): Possible involvement in the removal of CEL-modified proteins by macrophages", Biochemical and Biophysical Research Communications, Vol. 407, pp. 420-425 (March 12, 2011).
[249] 42. Reddy, S. et al., “N£-(carboxymethyl)lysine is a dominant advanced glycation end product (AGE) antigen in tissue proteins”, Biochemistry, Vol. 34, pp. 10872-10878 (August 1, 1995).
[250] 43. Naylor, R. M. et al., “Senescent cells: a novel therapeutic target for aging and age-related diseases”, Clinical Pharmacology & Therapeutics, Vol. 93(1), pp.105-116 (December 5, 2012). [251] 44. Katcher, H. L., “Studies that shed new light on aging", Biochemistry (Moscow), Vol. 78(9), pp. 1061-1070 (2013).
[252] 45. Fielding, R. A., etal., “Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences”, Journal of the American Medical Directors Association, Vol. 12(4), pp. 249-256 (May 2011).
[253] 46. Fu, M-X., et al., “The advanced glycation end product, Ne- (carboxymethyl)lysine, is a product of both lipid peroxidation and glycoxidation reactions”, The Journal of Biological Chemistry, Vol. 271, No. 17, pp. 9982-9986 (April 26, 1996).
[254] 47. Kudryashova, E. et al., “Satellite cell senescence underlies myopathy in a mouse model of limb-girdle muscular dystrophy 2H”, The Journal of Clinical Investigation, Vol. 122, No. 5, pp. 1764-1776 (May 2012).
[255] 48. Ratelade, J. et al., “Neuromyelitis optica IgG and natural killer cells produce NMO lesions in mice without myelin loss”, Acta Neuropathologica, Vol. 123, No. 6, pp. 861-872 (June 2012).
[256] 49. Vincent, T. et al., “Functional consequences of neuromyelitis optica- IgG astrocyte interactions on blood-brain barrier permeability and granulocyte recruitment”, The Journal of Immunology, Vol. 181, pp. 5730-5737 (2008).
[257] 50. Xu, M. et al., “Transplanted senescent cells induce an osteoarthritislike condition in mice”, Journals of Gerontology: Biological Sciences, Vol. 00, No. 00, 1-6 (2016).
[258] 51. Rayavarapu, S. et al., “Idiopathic inflammatory myopathies: pathogenic mechanisms of muscle weakness”, Skeletal Muscle, Vol. 3, 13 pages (June 2013).
[259] 52. Luessi, F., et al. “Neurodegeneration in multiple sclerosis: novel treatment strategies” Expert Rev. Neurother., Vol 9, pp.1061 -1077 (2012). [260] 53. Durieu, S. et al., “Subepithelial fibrosis and degradation of the bronchial extracellular matrix in cystic fibrosis”, American Journal of Respiratory and Critical Care Medicine, Vol. 158, pp. 580-588 (1998).
[261] 54. Shapiro, B. L. et al., “Premature senescence in cultured skin fibroblasts from subjects with cystic fibrosis", Science, Vol. 203, Issue 4386, pp. 1251-1253 (1979). '
[262] 55. Fischer, B. M. et al., “Increased expression of senescence markers in cystic fibrosis airways”, American Journal of Physiology Lung Cellular and Molecular Physiology, Vol. 304, pp. L394-L400 (2013).
[263] 56. Romagosa, C. etal., p16lnk4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors, Oncogene, Vol. 30, 2087-2097 (2011).
[264] 57. Thom, M. et al., “An investigation of the expression of G1 -phase cell cycle proteins in focal cortical dysplasia type I IB”, Journal of Neuropathology & Experimental Neurology, Vol. 66, No. 11 , pp. 1045-1055 (November 2007).
[265] 58. Baarine, M. et al., “ABCD1 deletion-induced mitochondrial dysfunction is corrected by SAHA: implication for adrenoleukodystrophy”, Journal of Neurochemistry, Vol. 133, No. 3, pp. 380-396 (2015).
[266] 59. Zhu, Y. et al., “The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs”, Aging Cell, vol. 14, pp. 644-658 (2015).
[267] 60. Roos, C.M. et al., “Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice”, Aging Cell (2016).
[268] 61. “Arthritis at a Glance 2016”, Centers for Disease Control and Prevention (2016).
[269] 62. Woolf, A. etal., “Burden of major musculoskeletal conditions", Bulletin of the World Health Organization, Vol. 81, No. 9, pp. 646-656 (2003). [270] 63. Pereira, D., et al., “The effect of osteoarthritis definition on prevalence and incidence estimates: a systematic review", Osteoarthritis and Cartilage, Vol. 19, pp. 1270-1285 (2011).
[271] 64. “Osteoarthritis”, available online at en.wikipedia.org/Osteoarthritis (December 13, 2016).
[272] 65. “What is osteoarthritis?”, National Institute of Arthritis and Musculoskeletal and Skin Diseases (November 2014).
[273] 66. Martin, J.A. etal., “Aging, articular cartilage chondrocyte senescence and osteoarthritis”, Biogerontology, Vol. 3, pp. 254-264 (2002).
[274] 67. Zhang, H.W. et al., “Recovery of function in osteoarthritic chondrocytes induced by p16lnk4a-specific siRNA in vitro", Rheumatology, Vol. 43, No. 5, pp. 555- 568 (2004).
[275] 68. Xu, M. et al., “Transplanted senescent cells induce an osteoarthritislike condition in mice”, Journals of Gerontology: Biological Sciences, Vol. 00, No. 00, pp. 1-6 (August 10, 2016).
[276] 69. Xu, Y-k. et al., “The role of MCP-1-CCR2 ligand-receptor axis in chondrocyte degradation and disease progress in knee osteoarthritis", Biological Research, Vol. 48, No. 64 (2015).
[277] 70. Jeon, O.H. et al., “Senescent cells and osteoarthritis: a painful connection", Journal of Clinical Investigation, Vol. 128, No. 4, pp. 1229-1237 (2018).
[278] 71. Jeon, O.H. et al., “Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment’, Nature Medicine, Vol. 23, pp. 775-781 (2017).

Claims

WHAT IS CLAIMED IS:
1. A method of treating or preventing the onset of osteoarthritis, comprising administering to a subject a composition comprising an anti-AGE antibody.
2. A method of treating or preventing the onset of osteoarthritis, comprising administering to a subject a composition comprising a first anti-AGE antibody and a second anti-AGE antibody; wherein the second anti-AGE antibody is different from the first anti-AGE antibody.
3. A method of treating a subject with osteoarthritis, comprising: a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating osteoarthritis; followed by a second administering of the anti-AGE antibody.
4. A composition for treating or preventing the onset of osteoarthritis, comprising
(a) a first anti-AGE antibody,
(b) a second anti-AGE antibody, and
(c) a pharmaceutically acceptable carrier, wherein the first anti-AGE antibody is different from the second anti-AGE antibody.
5. A method of treating or preventing the onset of osteoarthritis, comprising immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.
6. A method of treating a subject with osteoarthritis, comprising: administering a first vaccine comprising a first AGE antigen; and optionally, administering a second vaccine comprising a second AGE antigen; wherein the second AGE antigen is different from the first AGE antigen.
7. The method, use or composition of any of the preceding claims, wherein the composition further comprises a pharmaceutically acceptable carrier.
8. The method, use or composition of any of the preceding claims, wherein the subject is selected from the group consisting of humans, mice, rats, goats, sheep, cows, horses, dogs and cats.
9. The method, use or composition of any of the preceding claims, wherein the subject is a human.
10. The method, use or composition of any of the preceding claims, wherein the anti-AGE antibody is non-immunogenic to a species selected from the group consisting of humans, cats, dogs, horses, camels, alpaca, cattle, sheep, and goats.
11. The method, use or composition of any of the preceding claims, wherein the anti-AGE antibody is administered intravenously.
12. The method, use or composition of any of the preceding claims, wherein the anti-AGE antibody is administered intra-articularly into an affected joint.
13. The method, use or composition of any of the preceding claims, wherein the anti-AGE antibody binds an AGE antigen comprising at least one protein or peptide that exhibits AGE modifications selected from the group consisting of FFI, pyrraline, AFGP, ALI, carboxymethyllysine, carboxyethyllysine and pentosidine.
14. The method, use or composition of any of the preceding claims, wherein the first anti-AGE antibody and the second anti-AGE antibody bind AGE antigens comprising at least one protein or peptide that exhibit different AGE modifications selected from the group consisting of FFI , pyrraline, AFGP, ALI, carboxymethyllysine, carboxyethyllysine and pentosidine.
15. The method, use or composition of any of the preceding claims, wherein the composition is in unit dosage form.
16. The method, use or composition of any of the preceding claims, wherein the composition is in multidosage form
17. The method, use or composition of any of the preceding claims, wherein the composition is sterile.
18. The method, use or composition of any of the preceding claims, wherein the immunizing comprises administering a vaccine comprising an AGE antigen.
19. The method, use or composition of any of the preceding claims, wherein the vaccine comprises
(a) the AGE antigen,
(b) an adjuvant,
(c) optionally, a preservative, and
(d) optionally, an excipient.
20. The method, use or composition of any of the preceding claims, wherein the vaccine is administered in an amount effective to cause the immune system to produce antibodies to cells having AGE-modified proteins or peptides.
21. The method, use or composition of any of the preceding claims, wherein the AGE antigen is an AGE-modified protein or peptide selected from the group consisting of AGE-RNAse, AGE-human hemoglobin, AGE-albumin, AGE-BSA, AGE-human serum albumin, AGE-ovalbumin, AGE-low density lipoprotein, AGE- collagen IV, AGE-antithrombin III, AGE-calmodulin, AGE-insulin, AGE- ceruloplasmin, AGE-collagen, AGE-cathepsin B, AGE-albumin, AGE-crystallin, AGE- plasminogen activator, AGE-endothelial plasma membrane protein, AGE-aldehyde reductase, AGE-transferrin, AGE-fibrin, AGE-copper/zinc SOD, AGE-apo B, AGE- fibronectin, AGE-pancreatic ribose, AGE-apo A-l and II, AGE-hemoglobin, AGE- Na+/K+-ATPase, AGE-plasminogen, AGE-myelin, AGE-lysozyme, AGE- immunoglobulin, AGE-red cell Glu transport protein, AGE-P-N-acetyl hexominase, AGE-apo E, AGE-red cell membrane protein, AGE-aldose reductase, AGE-ferritin, AGE-red cell spectrin, AGE-alcohol dehydrogenase, AGE-haptoglobin, AGE-tubulin, AGE-thyroid hormone, AGE-fibrinogen, AGE- -microglobulin, AGE-sorbitol dehydrogenase, AGE-ai-antitrypsin, AGE-carbonate dehydratase, AGE-RNAse, AGE-low density lipoprotein, AGE-hexokinase, AGE-apo C-l, AGE-KLH and mixtures thereof.
22. The method, use or composition of any of the preceding claims, wherein the AGE antigen comprises at least one protein or peptide that exhibits AGE modifications selected from the group consisting of carboxymethyllysine, carboxyethyllysine, pentosidine, pyrraline, FFI, AFGP, and ALL
23. The method, use or composition of any of the preceding claims, wherein the vaccine is sterile.
24. The method, use or composition of any of the preceding claims, wherein the vaccine is in unit dosage form.
25. The method, use or composition of any of the preceding claims, wherein the vaccine is in multidosage form.
26. The method, use or composition of any of the preceding claims, further comprising testing the patient to determine if the osteoarthritis has been ameliorated, and repeating the immunizing, if necessary.
27. The method, use or composition of any of the previous claims, wherein the anti-AGE antibody comprises a protein or peptide comprising at least one amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, SEQ ID NO: 37, and SEQ ID NO: 39.
28. The method, use or composition of any of the previous claims, wherein the antibody comprises a protein or peptide comprising at least one amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, and SEQ ID NO: 28.
29. The method, use or composition of any of the previous claims, wherein the antibody comprises a heavy chain, and a light chain, wherein the heavy chain comprises an amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 17, SEQ ID NO: 29, SEQ ID NO: 31, and SEQ ID NO: 33, or the light chain comprises an amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 19, SEQ ID NO: 35, SEQ ID NO: 37, and SEQ ID NO: 39.
30. The method, use or composition of any of the previous claims, wherein the antibody comprises a heavy chain, and a light chain, wherein the heavy chain comprises an amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 17, SEQ ID NO: 29, SEQ ID NO: 31, and SEQ ID NO: 33, and the light chain comprises an amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 19, SEQ ID NO: 35, SEQ ID NO: 37, and SEQ ID NO: 39.
31. The method, use or composition of any of the previous claims, wherein the antibody comprises complementary determining region comprising at least one amino acid sequence having at least 90% sequence identity, preferably at least 95% sequence identity, more preferably at least 98% sequence identity, with an amino acid sequence selected from the group consisting of SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, and SEQ ID NO: 28, and the antibody is substantially non-immunogenic to a species selected from the group consisting of mice, rats, goats, sheep, cows, horses, dogs and cats.
32. The method, use or composition of any of the previous claims, wherein the antibody is conjugated to an agent that causes the destruction of AGE-modified cells.
33. The method, use or composition of any of the preceding claims, wherein the antibody is a humanized antibody.
34. The method, use or composition of any of the preceding claims, wherein the antibody is monoclonal.
35. The method, use or composition of any of the preceding claims, wherein the antibody is substantially non-immunogenic to humans.
36. The method, use or composition of any of the preceding claims, wherein the antibody has a rate of dissociation (ka) of at most 9 x 10-3 sec1.
37. The method, use or composition of any of the preceding claims, wherein the agent is selected from the group consisting of a toxin, a cytotoxic agent, magnetic nanoparticles, and magnetic spin-vortex discs.
38. The method, use or composition of any of the preceding claims, wherein the antibody includes constant regions which permit destruction of targeted cells by a subject’s immune system.
39. The method, use or composition of any of the preceding claims, wherein the antibody is a bi-specific antibody.
40. The method, use or composition of any of the preceding claims, wherein the AGE antigen comprises carboxymethyllysine conjugated with keyhole limpet hemocyanin (CML-KLH).
PCT/US2020/057539 2020-10-27 2020-10-27 Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens WO2022093195A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2020/057539 WO2022093195A1 (en) 2020-10-27 2020-10-27 Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2020/057539 WO2022093195A1 (en) 2020-10-27 2020-10-27 Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens

Publications (1)

Publication Number Publication Date
WO2022093195A1 true WO2022093195A1 (en) 2022-05-05

Family

ID=73598180

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/057539 WO2022093195A1 (en) 2020-10-27 2020-10-27 Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens

Country Status (1)

Country Link
WO (1) WO2022093195A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11518801B1 (en) 2017-12-22 2022-12-06 Siwa Corporation Methods and compositions for treating diabetes and diabetic complications
US11542324B2 (en) 2017-04-13 2023-01-03 Siwa Corporation Humanized monoclonal advanced glycation end-product antibody
US11833202B2 (en) 2016-02-19 2023-12-05 Siwa Corporation Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (AGE)
US11872269B2 (en) 2014-12-18 2024-01-16 Siwa Corporation Method and composition for treating sarcopenia
US11873345B2 (en) 2014-12-18 2024-01-16 Siwa Corporation Product and method for treating sarcopenia
US11958900B2 (en) 2016-04-15 2024-04-16 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4217344A (en) 1976-06-23 1980-08-12 L'oreal Compositions containing aqueous dispersions of lipid spheres
US4911928A (en) 1987-03-13 1990-03-27 Micro-Pak, Inc. Paucilamellar lipid vesicles
US4917951A (en) 1987-07-28 1990-04-17 Micro-Pak, Inc. Lipid vesicles formed of surfactants and steroids
US5702704A (en) 1991-12-20 1997-12-30 The Rockefeller University Antibodies to in vivo advanced glycosylation endproducts
US6380165B1 (en) 1997-09-19 2002-04-30 The Picower Institute For Medical Research Immunological advanced glycation endproduct crosslink
US6387373B1 (en) 1993-01-15 2002-05-14 Novavax, Inc. Vaccines containing paucilsmellar lipid vesicles as immunological adjuvants
WO2009143411A2 (en) 2008-05-23 2009-11-26 Siwa Corporation Methods, compositions and apparatus for facilitating regeneration
US20100226932A1 (en) 2006-02-22 2010-09-09 Novavax, Inc. Adjuvant and Vaccine Compositions
WO2016044252A2 (en) * 2014-09-19 2016-03-24 Siwa Corporation Anti-age antibodies for treating inflammation and auto-immune disorders
WO2020023532A1 (en) * 2018-07-23 2020-01-30 Siwa Corporation Methods and compositions for treating chronic effects of radiation and chemical exposure
WO2020041625A1 (en) * 2018-08-23 2020-02-27 Siwa Corporation Anti carboxymethyl lysine antibodies and ultrasound for removing age-modified cells

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4217344A (en) 1976-06-23 1980-08-12 L'oreal Compositions containing aqueous dispersions of lipid spheres
US4911928A (en) 1987-03-13 1990-03-27 Micro-Pak, Inc. Paucilamellar lipid vesicles
US4917951A (en) 1987-07-28 1990-04-17 Micro-Pak, Inc. Lipid vesicles formed of surfactants and steroids
US5702704A (en) 1991-12-20 1997-12-30 The Rockefeller University Antibodies to in vivo advanced glycosylation endproducts
US6387373B1 (en) 1993-01-15 2002-05-14 Novavax, Inc. Vaccines containing paucilsmellar lipid vesicles as immunological adjuvants
US6380165B1 (en) 1997-09-19 2002-04-30 The Picower Institute For Medical Research Immunological advanced glycation endproduct crosslink
US20100226932A1 (en) 2006-02-22 2010-09-09 Novavax, Inc. Adjuvant and Vaccine Compositions
WO2009143411A2 (en) 2008-05-23 2009-11-26 Siwa Corporation Methods, compositions and apparatus for facilitating regeneration
WO2016044252A2 (en) * 2014-09-19 2016-03-24 Siwa Corporation Anti-age antibodies for treating inflammation and auto-immune disorders
WO2020023532A1 (en) * 2018-07-23 2020-01-30 Siwa Corporation Methods and compositions for treating chronic effects of radiation and chemical exposure
WO2020041625A1 (en) * 2018-08-23 2020-02-27 Siwa Corporation Anti carboxymethyl lysine antibodies and ultrasound for removing age-modified cells

Non-Patent Citations (78)

* Cited by examiner, † Cited by third party
Title
"EMBUGenBank", Database accession no. AY631942
"GenBank", Database accession no. AJ245184
"The Pink Book, Epidemiology and Prevention of Vaccine-Preventable Diseases", September 2013, article "Vaccine Excipient & Media Summary"
"What is osteoarthritis?", November 2014, NATIONAL INSTITUTE OF ARTHRITIS AND MUSCULOSKELETAL AND SKIN DISEASES
AHMED, E. K. ET AL.: "Protein Modification and Replicative Senescence of WI-38 Human Embryonic Fibroblasts", AGING CELLS, vol. 9, 2010, pages 252,260
AHMED, E.K. ET AL.: "Protein Modification and Replicative Senescence of WI- 38 Human Embryonic Fibroblasts", AGING CELLS, vol. 9, 252, 260, 2010
AMELI, S. ET AL.: "Effect of Immunization With Homologous LDL and Oxidized LDL on Early Atherosclerosis in Hypercholesterolemic Rabbits", ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, vol. 16, 1996, pages 1074
ANDO, K. ET AL.: "Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation", BIOCHEM BIOPHYS RES COMMUN., vol. 258, 123, 125, 1999
ANDO, K. ET AL.: "Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation", BIOCHEM BIOPHYS RES COMMUN.,, vol. 258, 123, 125, 1999
ANDO, K. ET AL.: "Membrane Proteins of Human Erythrocytes Are Modified by Advanced Glycation End Products during Aging in the Circulation", BIOCHEM. BIOPHYS. RES. COMMUN., vol. 258, 1999, pages 123,125
AROIAN R ET AL.: "Pore-Forming Toxins and Cellular Non-Immune Defenses (CNIDs", CURRENT OPINION IN MICROBIOLOGY, vol. 10, 2007, pages 57 - 61, XP005884635
ARTHRITIS AT A GLANCE 2016, 2016
BAARINE, M. ET AL.: "ABCD1 deletion-induced mitochondrial dysfunction is corrected by SAHA: implication for adrenoleukodystrophy", JOURNAL OF NEUROCHEMISTRY, vol. 133, no. 3, 2015, pages 380 - 396
BAKER, D. J. ET AL.: "Clearance of p16lnk4a-positive senescent cells delays ageing-associated disorders", NATURE, vol. 479, 2011, pages 232 - 236, XP055074325, DOI: 10.1038/nature10600
BEIER, K. C. ET AL.: "Master Switches of T-cell Differentiation", EUR. RESPIR. J., vol. 29, 2007, pages 804 - 12
BIERHAUS A: "AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept", CARDIOVASC RES, vol. 37, no. 3, 1998, pages 586 - 600, XP055399820, DOI: 10.1016/S0008-6363(97)00233-2
BIERHAUS, A.: "AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept", CARDIOVASC. RES., vol. 37, no. 3, 1998, pages 586 - 600, XP055399820, DOI: 10.1016/S0008-6363(97)00233-2
BLAHD, W., SARCOPENIA WITH AGING, 3 August 2014 (2014-08-03), Retrieved from the Internet <URL:www.webmd.com/healthy-aging/sarcopenia-with-aging>
BUSKAS, T. ET AL.: "Immunotherapy for Cancer: Synthetic Carbohydrate-based Vaccines", CHEM. COMMUN., vol. 28, no. 36, 2009, pages 5335 - 349
CHEADLE, E. J. ET AL.: "Bugs as Drugs for Cancer", IMMUNOLOGY, vol. 107, 2002, pages 10 - 19
CML (N-EPSILON-(CARBOXYMETHYL)LYSINE) ASSAYS AND REAGENTS, 15 December 2014 (2014-12-15), Retrieved from the Internet <URL:atwww.cellbiolabs.com/cml-assays>
CML-BSA PRODUCT DATA SHEET, 2010, Retrieved from the Internet <URL:www.cellbiolabs.com/sites/default/files/STA-314-cml-bsa.pdf>
COLER, R. N. ET AL.: "Development and Characterization of Synthetic Glucopyranosyl Lipid Adjuvant System as a Vaccine Adjuvant", PLOS ONE, vol. 6, no. 1, 2011, pages e16333, XP055030257, DOI: 10.1371/journal.pone.0016333
CRUZ-JENTOFT, A. J. ET AL.: "Sarcopenia: European consensus on definition and diagnosis", AGE AND AGEING, vol. 39, 13 April 2010 (2010-04-13), pages 412 - 423, XP009167538, DOI: 10.1093/ageing/afq034
DE GROOT, K. ET AL.: "Vascular Endothelial Damage and Repair in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis", ARTHRITIS AND RHEUMATISM, vol. 56, no. 11, 2007, pages 3847,3847
DURIEU, S. ET AL.: "Subepithelial fibrosis and degradation of the bronchial extracellular matrix in cystic fibrosis", AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, vol. 158, 1998, pages 580 - 588
FERRACCIOLI, G. ET AL.: "Interleukin-1β and Interleukin-6 in Arthritis Animal Models: Roles in the Early Phase of Transition from Acute to Chronic Inflammation and Relevance for Human Rheumatoid Arthritis", MOL MED, vol. 16, no. 11-12, November 2010 (2010-11-01), pages 552 - 557
FIELDING, R. A. ET AL.: "Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences", JOURNAL OF THE AMERICAN MEDICAL DIRECTORS ASSOCIATION, vol. 12, no. 4, May 2011 (2011-05-01), pages 249 - 256
FISCHER, B. M. ET AL.: "Increased expression of senescence markers in cystic fibrosis airways", AMERICAN JOURNAL OF PHYSIOLOGY LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, vol. 304, 2013, pages L394 - L400
FLINT, S. J. ET AL.: "Principles of Virology", 2000, ASM PRESS
FREUND, A.: "Inflammatory networks during cellular senescence: causes and consequences", TRENDS MOL MED, vol. 16, no. 5, May 2010 (2010-05-01), pages 238 - 46, XP027048786
FU, M-X. ET AL.: "The advanced glycation end product, Nε-(carboxymethyl)lysine, is a product of both lipid peroxidation and glycoxidation reactions", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, no. 17, 26 April 1996 (1996-04-26), pages 9982 - 9986, XP055171871, DOI: 10.1074/jbc.271.17.9982
GAO, S. H. ET AL.: "Monoclonal antibody humanness score and its applications", BMC BIOTECHNOLOGY, vol. 13, 5 July 2013 (2013-07-05), pages 55, XP021156542, DOI: 10.1186/1472-6750-13-55
JEON, O.H. ET AL.: "Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment", NATURE MEDICINE, vol. 23, 2017, pages 775 - 781
JEON, O.H. ET AL.: "Senescent cells and osteoarthritis: a painful connection", JOURNAL OF CLINICAL INVESTIGATION, vol. 128, no. 4, 2018, pages 1229 - 1237
JIANG, Z-H. ET AL.: "Synthetic vaccines: the role of adjuvants in immune targeting", CURRENT MEDICINAL CHEMISTRY, vol. 10, no. 15, 2003, pages 1423 - 39, XP009017277, DOI: 10.2174/0929867033457340
KAJSTURA, J. ET AL.: "Myocite Turnover in the Aging Human Heart", CIRC. RES., vol. 107, no. 11, 2010, pages 1374 - 86
KATCHER, H. L.: "Studies that shed new light on aging", BIOCHEMISTRY (MOSCOW), vol. 78, no. 9, 2013, pages 1061 - 1070, XP035358815, DOI: 10.1134/S0006297913090137
KEYHOLE LIMPET HEMOCYANIN, 18 April 2014 (2014-04-18), Retrieved from the Internet <URL:en.wikipedia.org/wiki/Keyhole_limpet_hemocyanin>
KIRSTEIN, M. ET AL.: "Receptor-specific Induction of Insulin-like Growth Factor I in Human Monocytes by Advanced Glycosylation End Product-modified Proteins", J. CLIN. INVEST., vol. 90, no. 439, 1992, pages 439 - 440
KUDRYASHOVA, E. ET AL.: "Satellite cell senescence underlies myopathy in a mouse model of limb-girdle muscular dystrophy 2H", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 122, no. 5, May 2012 (2012-05-01), pages 1764 - 1776
LINDSEY, J. B. ET AL.: "Receptor For Advanced Glycation End-Products (RAGE) and soluble RAGE (sRAGE): Cardiovascular Implications", DIABETES VASCULAR DISEASE RESEARCH, vol. 6, no. 1, 2009, pages 7 - 14, XP055494933, DOI: 10.3132/dvdr.2009.002
LUESSI, F. ET AL.: "Neurodegeneration in multiple sclerosis: novel treatment strategies", EXPERT REV. NEUROTHER., vol. 9, 2012, pages 1061 - 1077, XP055337068, DOI: 10.1586/ern.12.59
MANESSO, E. ET AL.: "Dynamics of P-Cell Turnover: Evidence for β-Cei) Turnover and Regeneration from Sources of (3-Cells other than 13-cell Replication in the HIP Rat", AM. J. PHYSIOL. ENDOCRINOL. METAB., vol. 297, 2009, pages E323,E324
MARTIN, J.A. ET AL.: "Aging, articular cartilage chondrocyte senescence and osteoarthritis", BIOGERONTOLOGY, vol. 3, 2002, pages 254 - 264
MERA, K. ET AL.: "An autoantibody against N£-(carboxyethyl)lysine (CEL): Possible involvement in the removal of CEL-modified proteins by macrophages", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 407, 12 March 2011 (2011-03-12), pages 420 - 425
MURPHY, J. F.: "Trends in cancer immunotherapy", CLINICAL MEDICAL INSIGHTS: ONCOLOGY, vol. 14, no. 4, 2010, pages 67 - 80
NAYLOR, R. M.: "Senescent cells: a novel therapeutic target for aging and age-related diseases", CLINICAL PHARMACOLOGY & THERAPEUTICS, vol. 93, no. 1, 5 December 2012 (2012-12-05), pages 105 - 116, XP002710105, DOI: 10.1038/clpt.2012.193
NELSON, G.: "A senescent cell bystander effect: senescence-induced senescence", AGING CELL, vol. 11, 2012, pages 345 - 349, XP055625700, DOI: 10.1111/j.1474-9726.2012.00795.x
OSTEOARTHRITIS, 13 December 2016 (2016-12-13), Retrieved from the Internet <URL:en.wikipedia.org/Osteoarthritis>
PEREIRA, D. ET AL.: "The effect of osteoarthritis definition on prevalence and incidence estimates: a systematic review", OSTEOARTHRITIS AND CARTILAGE, vol. 19, 2011, pages 1270 - 1285, XP028312365, DOI: 10.1016/j.joca.2011.08.009
RATELADE, J.: "Neuromyelitis optica IgG and natural killer cells produce NMO lesions in mice without myelin loss", ACTA NEUROPATHOLOGICA, vol. 123, no. 6, June 2012 (2012-06-01), pages 861 - 872, XP035060625, DOI: 10.1007/s00401-012-0986-4
RAYAVARAPU, S. ET AL.: "Idiopathic inflammatory myopathies: pathogenic mechanisms of muscle weakness", SKELETAL MUSCLE, vol. 3, June 2013 (2013-06-01), pages 13, XP021153799, DOI: 10.1186/2044-5040-3-13
RAYESS, H: "Cellular senescence and tumor suppressor gene", INT J CANCER, vol. 130, 2012, pages 1715 - 1725, XP055282399, DOI: 10.1002/ijc.27316
REDDY, S. ET AL.: "N£-(carboxymethyl)lysine is a dominant advanced glycation end product (AGE) antigen in tissue proteins", BIOCHEMISTRY, vol. 34, 1 August 1995 (1995-08-01), pages 10872 - 10878, XP002088366, DOI: 10.1021/bi00034a021
ROLL, P. ET AL.: "Anti-CD20 Therapy in Patients with Rheumatoid Arthritis", ARTHRITIS & RHEUMATISM, vol. 58, no. 6, 2008, pages 1566 - 1575, XP055030411, DOI: 10.1002/art.23473
ROLLAND, Y. ET AL.: "Sarcopenia: its assessment, etiology, pathogenesis, consequences and future perspectives", J. NUTR. HEALTH AGING, vol. 12, no. 7, 2008, pages 433 - 450
ROMAGOSA, C. ET AL.: "p16lnk4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors", ONCOGENE, vol. 30, 2011, pages 2087 - 2097
ROOS, C.M. ET AL.: "Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice", AGING CELL, 2016
SAMAN AHMAD ET AL: "Preferential recognition of epitopes on AGE-IgG by the autoantibodies in rheumatoid arthritis patients", HUMAN IMMUNOLOGY, vol. 74, no. 1, 1 January 2013 (2013-01-01), US, pages 23 - 27, XP055237572, ISSN: 0198-8859, DOI: 10.1016/j.humimm.2012.10.008 *
SARCOPENIA, 14 November 2014 (2014-11-14), Retrieved from the Internet <URL:en.wikipedia.org/wiki/Sarcopenia>
SCHMIDLIN H. ET AL.: "New Insights in the Regulation of Human B Cell Differentiation", TRENDS IMMUNOL., vol. 30, no. 6, 2009, pages 277 - 85, XP026171863, DOI: 10.1016/j.it.2009.03.008
SHAPIRO, B. L. ET AL.: "Premature senescence in cultured skin fibroblasts from subjects with cystic fibrosis", SCIENCE, vol. 203, no. 4386, 1979, pages 1251 - 1253
THOM, M. ET AL.: "An investigation of the expression of G1-phase cell cycle proteins in focal cortical dysplasia type IIB", JOURNAL OF NEUROPATHOLOGY & EXPERIMENTAL NEUROLOGY, vol. 66, no. 11, November 2007 (2007-11-01), pages 1045 - 1055
VINCENT, T. ET AL.: "Functional consequences of neuromyelitis optica-IgG astrocyte interactions on blood-brain barrier permeability and granulocyte recruitment", THE JOURNAL OF IMMUNOLOGY, vol. 181, 2008, pages 5730 - 5737
VIRELLA, G. ET AL.: "Autoimmune Response to Advanced Glycosylation End-Products of Human LDL", JOURNAL OF LIPID RESEARCH, vol. 44, 2003, pages 487 - 493, XP055494935, DOI: 10.1194/jlr.M200370-JLR200
VLASSARA ET AL.: "High-affinity-receptor-mediated Uptake and Degradation of Glucose-modified Proteins: A Potential Mechanism for the Removal of Senescent Macromolecules", PROC. NATL. ACAD. SCI. USAI, vol. 82, 1985, pages 5588,5591
VLASSARA, H. ET AL.: "Advanced Glycosylation Endproducts on Erythrocyte Cell Surface Induce Receptor-Mediated Phagocytosis by Macrophages", J. EXP. MED., vol. 166, 539, 545, 1987, pages 539,545
VLASSARA, H. ET AL.: "High-affinity-receptor-mediated Uptake and Degradation of Glucose-modified Proteins: A Potential Mechanism for the Removal of Senescent Macromolecules", PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 5588,5591
VOGEL, F. R. ET AL.: "A compendium of vaccine adjuvants and excipients", PHARMACEUTICAL BIOTECHNOLOGY, vol. 6, 1995, pages 141 - 228
WHAT IS SARCOPENIA?, 2014, Retrieved from the Internet <URL:www.iofbonehealth.org/what-sarcopenia>
WOOLF, A. ET AL.: "Burden of major musculoskeletal conditions", BULLETIN OF THE WORLD HEALTH ORGANIZATION, vol. 81, no. 9, 2003, pages 646 - 656
XU, M. ET AL.: "Transplanted senescent cells induce an osteoarthritis-like condition in mice", JOURNALS OF GERONTOLOGY: BIOLOGICAL SCIENCES, vol. 00, no. 00, 10 August 2016 (2016-08-10), pages 1 - 6
XU, M.: "Transplanted senescent cells induce an osteoarthritis-like condition in mice", JOURNALS OF GERONTOLOGY: BIOLOGICAL SCIENCES, vol. 00, no. 00, 2016, pages 1 - 6
XU, Y-K. ET AL.: "The role of MCP-1-CCR2 ligand-receptor axis in chondrocyte degradation and disease progress in knee osteoarthritis", BIOLOGICAL RESEARCH, vol. 48, no. 64, 2015
ZHANG, H.W. ET AL.: "Recovery of function in osteoarthritic chondrocytes induced by p16lnk4a-specific siRNA in vitro", RHEUMATOLOGY, vol. 43, no. 5, 2004, pages 555 - 568, XP002418725, DOI: 10.1093/rheumatology/keh127
ZHANG, H.W. ET AL.: "Recovery of function in osteoarthritic chondrocytes induced by pl61nk4a-specific siRNA in vitro", RHEUMATOLOGY, vol. 43, no. 5, 2004, pages 555 - 568, XP002418725, DOI: 10.1093/rheumatology/keh127
ZHU, Y. ET AL.: "The Achilles' heel of senescent cells: from transcriptome to senolytic drugs", AGING CELL, vol. 14, 2015, pages 644 - 658, XP055342891, DOI: 10.1111/acel.12344

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11872269B2 (en) 2014-12-18 2024-01-16 Siwa Corporation Method and composition for treating sarcopenia
US11873345B2 (en) 2014-12-18 2024-01-16 Siwa Corporation Product and method for treating sarcopenia
US11833202B2 (en) 2016-02-19 2023-12-05 Siwa Corporation Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (AGE)
US11958900B2 (en) 2016-04-15 2024-04-16 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
US11542324B2 (en) 2017-04-13 2023-01-03 Siwa Corporation Humanized monoclonal advanced glycation end-product antibody
US11518801B1 (en) 2017-12-22 2022-12-06 Siwa Corporation Methods and compositions for treating diabetes and diabetic complications

Similar Documents

Publication Publication Date Title
US10858449B1 (en) Methods and compositions for treating osteoarthritis
US20220175916A1 (en) Methods and compositions for treating chronic effects of radiation and chemical exposure
US11873345B2 (en) Product and method for treating sarcopenia
US20210253737A1 (en) Methods and compositions for treating disease-related cachexia
AU2017219749B2 (en) Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (AGE)
US10961321B1 (en) Methods and compositions for treating pain associated with inflammation
WO2022093195A1 (en) Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens
JP2018535953A (en) Anti-AGE antibody and method of use thereof
US20230181730A1 (en) Methods of treating infections
CA3057829A1 (en) Anti-age antibodies for treating neurodegenerative disorders
US11518801B1 (en) Methods and compositions for treating diabetes and diabetic complications
WO2021247397A2 (en) Methods and compositions for enhancing the immune system
WO2023023654A1 (en) Methods and compositions for treating fibrotic diseases
US20240000930A1 (en) Methods and compositions for treating kidney diseases
WO2024102157A1 (en) Methods and compositions for treating diabetes and diabetic complications
US20160106837A1 (en) Combination of cd37 antibodies with chlorambucil

Legal Events

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

Ref document number: 20812462

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20812462

Country of ref document: EP

Kind code of ref document: A1