WO2021222758A1 - Methods of treating infections - Google Patents

Methods of treating infections Download PDF

Info

Publication number
WO2021222758A1
WO2021222758A1 PCT/US2021/030184 US2021030184W WO2021222758A1 WO 2021222758 A1 WO2021222758 A1 WO 2021222758A1 US 2021030184 W US2021030184 W US 2021030184W WO 2021222758 A1 WO2021222758 A1 WO 2021222758A1
Authority
WO
WIPO (PCT)
Prior art keywords
age
antibody
seq
composition
infection
Prior art date
Application number
PCT/US2021/030184
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 IL297799A priority Critical patent/IL297799A/en
Priority to CN202180047404.4A priority patent/CN115996954A/en
Priority to CA3177449A priority patent/CA3177449A1/en
Priority to JP2022566616A priority patent/JP2023524098A/en
Priority to AU2021264007A priority patent/AU2021264007A1/en
Priority to US17/922,264 priority patent/US20230181730A1/en
Priority to EP21731330.3A priority patent/EP4143238A1/en
Publication of WO2021222758A1 publication Critical patent/WO2021222758A1/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Viruses are infectious agents that contain genetic material in the form of DNA or RNA within a protein coat known as a capsid.
  • a complete virus particle of genetic material within a capsid is referred to as a virion.
  • Some viruses have a viral envelope around the capsid.
  • the viral envelope is typically composed of portions of cell membranes derived from a host cell and may also include viral glycoproteins. Examples of viruses that include a viral envelope include herpesvirus, poxvirus, hepadnavirus, asfivirus, flavivirus, alphavirus, togavirus, coronavirus, hepatitis D, orthomyxovirus, paramyxovirus, rhabdovirus, bunyavirus, filovirus and retroviruses.
  • a viral infection occurs when a host organism is introduced to a pathogenic virus that replicates inside the cells of the organism.
  • Viral replication is the process by which viruses take over or “hijack” the host cells to obtain energy and manufacture new viruses to facilitate the spread of the viral infection, both within the host and to new hosts.
  • Rapidly replicating viruses are of particular concern because of their ability to quickly spread and infect new hosts. Widespread viral infections can progressively grow into outbreaks, epidemics and pandemics. Rapidly replicating viruses can also overwhelm the host organism, which may lead to organ damage or death.
  • Examples of rapidly replicating viruses include influenza, such as influenza A virus subtype H5N1, coronaviruses, such as Middle East respiratory syndrome-related coronavirus (MERS-CoV) and severe acute respiratory syndrome-related coronavirus (SARS- CoV and SARS-CoV-2), and Ebola virus. These viruses have caused epidemics of the diseases known as the bird flu, MERS (Middle-East respiratory syndrome) and SARS (severe acute respiratory syndrome), and the pandemic of the disease known as COVID-19.
  • MERS Middle East respiratory syndrome-related coronavirus
  • SARS-CoV-2 severe acute respiratory syndrome-related coronavirus
  • COVID-19 the pandemic of the disease known as COVID-19.
  • Pulmonary pathology in early-phase COVID-19 pneumonia has shown exudative and proliferative phases of acute lung injury (edema, inflammatory infiltrates, pneumocyte hyperplasia) prior to the development of any respiratory symptoms.
  • acute lung injury edema, inflammatory infiltrates, pneumocyte hyperplasia
  • ARDS acute respiratory distress syndrome
  • ARDS has taken center stage as the primary cause of death in the global COVID-19 crisis.
  • the “cytokine storm” is emerging as a key mechanism leading to patient deterioration and death. Cytokine storm and ARDS has been associated with death from other viral infections, including SARS (SARS-COV), influenza viruses and the Spanish flu virus which caused the 1918 pandemic.
  • SARS SARS
  • influenza viruses influenza viruses
  • Spanish flu virus which caused the 1918 pandemic.
  • Cytokine storm is associated with marked morbidity and mortality in patients presenting during the COVID-19 pandemic.
  • the overwhelming progression to pulmonary failure is the hallmark vicious cycle overwhelming worldwide healthcare resources.
  • M. tuberculosis causes the formation of hard nodules or tubercles in the lungs, parasitizes macrophages by blocking the phagosome-lysosome fusion, a process called phagosome maturation arrest, and by replicating inside the phagosome (Vergne I, et al. Cell Biology of Mycobacterium tuberculosis Phagosome, Ann Rev Cell Dev Biol., Vol. 20, 367-94 (2004)).
  • P. tuberculosis causes the formation of hard nodules or tubercles in the lungs, parasitizes macrophages by blocking the phagosome-lysosome fusion, a process called phagosome maturation arrest, and by replicating inside the phagosome (Vergne I, et al. Cell Biology of Mycobacterium tuberculosis Phagosome, Ann Rev Cell Dev Biol., Vol. 20, 367-94 (2004)).
  • aeruginosa colonizes the lungs of patients with cystic fibrosis and produces biofilms, alginates, and specific lipid A modifications, which allow the bacteria to escape immune response and cause severe chronic inflammation (Moskowitz SM, et al. The Role of Pseudomonas Lipopolysaccharide in Cystic Fibrosis Airway Infection, Subcell Biochem., Vol. 53, 241-53 (2010)). Production of biofilms by Haemophilus influenzae, Streptococcus pneumoniae, and other bacteria, has been linked to chronic otitis media in pediatric patients (Hall-Stoodley L, et al. Direct Detection of Bacterial Biofilms on the Middle-Ear Mucosa of Children With Chronic Otitis Media, JAMA, Vol. 256, No. 2, 202-11 (2006)).
  • Plasmodium Leishmania, Trypanosoma and Toxoplasma. Plasmodium, the agent causing malaria, replicates and accumulates inside erythrocytes, provoking cell rupture and dissemination of the agent, while the main sites of sequestration of the infected erythrocytes containing the trophozoites, schizonts and gametocytes of the parasite have been shown to be the lung, spleen, and adipose tissue, but also the brain, skin, bone marrow, and skeletal and cardiac muscle (Franke-Fayard B, etal. Sequestration and Tissue Accumulation of Human Malaria Parasites: Can We Learn Anything from Rodent Models of Malaria?, PLoS Pathogens, Vol.
  • fungi are parasites on plants, animals (including humans), and other fungi. Fungus may invade tissue and can cause a disease. Some fungi may cause serious disease in humans. Fungi can attack eyes, nails, hair, and especially skin.
  • One common fungal infection is Valley Fever, which is caused by Coccidioides immitis (CF). Valley Fever usually occurs due to inhalation of the arthroconidial spores of CF after soil disruption. Once inhaled, the spores enter the alveoli and enlarge in size to become spherules, and internal septations develop. Septations develop and form endospores within the spherule. The rupture of the spherules releases the endospores, which in turn repeat the cycle and spread the infection to adjacent tissues within the body.
  • CF Coccidioides immitis
  • 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 b-galactosidase. Senescence begins with damage or stress (such as overstimulation by growth factors) of cells.
  • AGEs Advanced glycation end-products
  • AGEs also referred to as AGE-modified proteins or peptides, or glycation end-products
  • AGE-modified proteins or peptides or glycation end-products
  • Maho K. et ai, 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.
  • 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 inflammation, 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 AGEs and senescence is well known in the art. See, for example, Gruber, L. (WO 2009/143411 , 26 Nov. 2009), Ando, K. etal. (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)), Ahmed, E.K. et al. (“Protein Modification and Replicative Senescence of WI-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 end- products is associated with senescence and lack of function.
  • 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 hyperprol iterative state. However, there has been some ambiguity in this regard, as some tumors show overexpression of p16, while others 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. etal., 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).
  • SASP senescence-associated secretory phenotype
  • Autoimmune diseases such as Crohn’s disease and rheumatoid arthritis, are associated with chronic inflammation (Ferraccioli, G. etal.
  • 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.
  • Senescent cells also upregulate genes with roles in inflammation including IL-1 b, IL-8, ICAM1, TNFAP3, ESM1 and CCL2 (Burton, D.G.A. etal., “Microarray analysis of senescent vascular smooth muscle cells: a link to atherosclerosis and vascular calcification”, Experimental Gerontology, Vol. 44, No. 10, pp. 659-665 (October 2009)). 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. etal., Cellular senescence and tumor suppressor gene p16, IntJ 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 an infection comprising administering to a subject a composition comprising an anti-AGE antibody.
  • the invention is a method of treating an infection 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 an infection comprising a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating the infection; 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 an infection.
  • the invention is a composition comprising an anti-AGE antibody for use in treating an infection.
  • the invention is a composition for treating an infection 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 an infection comprising immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.
  • the invention is a method of treating an infection 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 an infection.
  • the invention is a composition comprising an AGE antigen for use in treating an infection.
  • peptide means a molecule composed of 2-50 amino acids.
  • the term “protein” means a molecule composed of more than 50 amino acids.
  • the terms “advanced glycation end-product”, “AGE”, “AGE-modified protein”, “AGE-modified 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)-1 H-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 p10 i n k4 a or expression of senescence-associated b-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, CA), 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.
  • FIG. 2 is a graph of the number of events and the fluorescence intensity for antibodies bound to cells having various multiplicities of infection (MOI) of influenza virus.
  • FIG. 3 is a graph of the number of events and the fluorescence intensity for antibodies bound to cells for various multiplicities of infection (MOI) of influenza virus.
  • Viruses must reprogram the host cell metabolism to increase the supply of nutrients, energy and metabolites that are necessary for replication. Viral control over host-cell metabolism involves upregulation of a carbon source, typically glucose or glutamine, and a redirection of these carbon supplies to metabolic pathways (Mayer, K.A. eta!., “Hijacking the supplies: metabolism as a novel facet of virus-host interaction”, Frontiers in Immunology, Vol. 10, Article 1533, 12 pages (2019)).
  • a carbon source typically glucose or glutamine
  • Viruses obtain energy from the additional glucose through glycolysis (the enzymatic break down of glucose) and glycosylation (the enzymatic process that attaches glycansto proteins) (“Novel Coronavirus COVID-19”, Moleculin Biotech, available online at www.moleculin.com/covid-19/ (accessed April 28, 2020)).
  • glycolysis the enzymatic break down of glucose
  • glycosylation the enzymatic process that attaches glycansto proteins
  • Enhanced glycolysis is also observed in cancer and oncogenic viruses and is known as aerobic glycolysis or the Warburg effect (Yu, L etal., “Oncogenic virus-induced aerobic glycolysis and tumorigenesis”, Journal of Cancer, Vol. 9, No. 20, pp. 3699-3706 (2016)).
  • COVID-19 has been shown to rely on both glycolysis and glycosylation to fuel its growth.
  • the characteristic spikes that surround coronaviruses such as SARS- CoV-2 are glycoproteins, which are formed by glycosylation.
  • Multiple studies have shown that disrupting glycolysis and glycosylation is effective in stopping viruses like coronavirus (Moleculin Biotech).
  • the glucose decoy 2-deoxy-D- glucose (2-DG) has been shown to block glycolysis and completely prevent SARS- CoV-2 replication in human cells (Bojkova, D.
  • ROS Reactive oxygen species
  • Antibodies that bind to advanced glycation end-products have been shown to effectively treat age-related diseases such as sarcopenia (US 9,161 ,810) and metastatic cancer (WO 2017/143073) by binding and removing AGE- modified cells, such as senescent cells.
  • Anti-AGE antibodies may be similarly used to bind cells that have been AGE-modified as a result of increased metabolic activity due to viral infection, such as highly glycolytic and glycoxidative cells.
  • Warburg effect is observed in cells infected with intracellular bacteria, such as tuberculosis infections (Shi, L et al., “Biphasic dynamics of macrophage inmmunometabolism during Mycobacterium tuberculosis infection” mBio, Vol. 10,
  • the enhanced glycolysis would also be expected to occur in parasite infections, as parasite reproduction inside cells requires energy (see Traore, K. et al., “Do advanced glycation end- products play a role in malaria susceptibility?”, Parasite, Vol. 23, No. 15, pp. 1-10 (2016)).
  • Malarial parasites use aerobic glycolysis, and may produce elevated levels of carboxymethyllysine. (Sturm, et al. “Mitochondrial ATP synthase is dispensable in blood-stage Plasmodium berghei rodent malaria but essential in the mosquito phase”, PNAS, Vol. 112, No. 33, pp. 10216-10223 (2015)).
  • Anti-AGE antibodies may also be used to bind AGE-modified proteins or peptides present on a viral envelope.
  • Glycoproteins present in the viral envelope may be glycated due to the elevated level of reactive oxygen species and oxidative stress that occur during viral replication. The glycation of viral envelope glycoproteins forms AGEs, such as carboxymethyllysine, that will be recognized by anti-AGE antibodies.
  • AGEs such as carboxymethyllysine
  • the inflammatory response observed in viral infections also indicates that anti-AGE antibodies would be effective therapies against viral infections.
  • the inflammatory response that is observed in viral infections is similarly observed with bacterial infections and parasitic infections. Interferons, particularly inflammatory cytokines, are produced during viral infections by the immune system (Eisenreich,
  • the cytokine storm is a systemic inflammatory response that is characterized by the release of Inflammatory cytokines. Senescent cells are known to secrete inflammatory factors and reactive oxygen species as part of the senescence-associated secretory phenotype (SASP), and the removal of these AGE-modified cells has been used to treat inflammation and auto-immune disorders (WO 2016/044252). The role of the cytokine storm observed in COVID-19 suggests that removal of AGE-modified cells would be similarly effective at reducing the inflammatory aspects of infections.
  • the increased oxidative state resulting from the amplified metabolic activity during viral, bacterial, parasitic and fungal replication combined with the increased inflammatory environment from the cytokine storm indicates that AGE-modified cells are an appropriate therapeutic target for treating viral, bacterial, parasitic and fungal infections.
  • the targeted removal of AGE-modified cells will reduce oxidative damage and reduce inflammation.
  • the present invention uses enhanced clearance of cells expressing AGE-modified proteins or peptides (AGE-modified cells) to treat a viral infection.
  • the present invention also includes enhanced clearance of cells expressing AGE-modified proteins or peptides (AGE-modified cells) to treat a bacterial, parasitic or fungal invention. This may be accomplished by administering anti-AGE antibodies to a subject.
  • the anti-AGE antibodies may also bind to AGEs present on the viral envelope to remove virions and viruses or AGEs present on bacteria, parasites or fungi.
  • 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 AGE-modified cells.
  • the process of AGE-modified cell removal or destruction allows vaccination against AGE-modified proteins or peptides of a cell to be used to treat a viral infection, bacterial infection or parasitic infection.
  • Vaccination against AGE-modified proteins or peptides also allows the immune system to target AGEs present on the viral envelope of virions and viruses, as well as bacteria and parasites.
  • 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 etai).
  • the antibody may bind to one or more AGE-modified proteins or peptides having an AGE modification such as FFI, pyrraline, AFGP, ALI, carboxymethyllysine, carboxyethyllysine and pentosidine, and mixtures of such antibodies.
  • 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), camelized (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.
  • 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 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), 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 10 3 , 7 x 10 3 or 6 x 10 3 (sec 1 ).
  • 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.
  • 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 antibody- dependent 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. et al., “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 are Y-shaped proteins composed of two heavy chains and two light chains.
  • the two arms of the Y shape form the fragment antigen-binding (Fab) region while the base or tail of the Y shape forms the fragment crystallizable (Fc) region of the antibody.
  • Antigen binding occurs at the terminal portion of the fragment antigen-binding region (the tips of the arms of the Y shape) at a location referred to as the paratope, which is a set of complementarity determining regions (also known as CDRs or the hypervariable region).
  • the complementarity determining regions vary among different antibodies and gives a given antibody its specificity for binding to a given antigen.
  • the fragment crystallizable region of the antibody determines the result of antigen binding and may interact with the immune system, such as by triggering the complement cascade or initiating antibody-dependent cell-mediated cytotoxicity (ADCC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • 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 a humanized anti-AGE antibody 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 a humanized anti-AGE antibody 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.
  • 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 preparation of non-human anti-AGE 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 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.
  • anti-AGE antibodies may be obtained by humanizing murine monoclonal anti-AGE antibodies.
  • Murine monoclonal anti-AGE antibodies have the heavy chain protein sequence shown in SEQ ID NO: 47 (the protein sequence of the variable domain is shown in SEQ ID NO: 52) and the light chain protein sequence shown in SEQ ID NO: 57 (the protein sequence of the variable domain is shown in SEQ ID NO: 62).
  • a preferred humanized heavy chain may have the protein sequence shown in SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 or SEQ ID NO: 51 (the protein sequences of the variable domains of the humanized heavy chains are shown in SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55 and SEQ ID NO: 56, respectively).
  • a preferred humanized light chain may have the protein sequence shown in SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60 or SEQ ID NO: 61 (the protein sequences of the variable domains of the humanized light chains are shown in SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65 and SEQ ID NO: 66, respectively).
  • a humanized anti-AGE monoclonal antibody is composed a heavy chain having a protein sequence selected from the group consisting of SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51 and a light chain having a protein sequence selected from the group consisting of SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60 and SEQ ID NO: 61.
  • Humanized monoclonal anti-AGE antibodies composed of these protein sequences may have better binding and/or improved activation of the immune system, resulting in greater efficacy.
  • 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 Camelus dromedarius and Camelus bactrianus
  • llamas Lama glama, Lama pacos and Lama vicugna
  • alpacas Vicugna pacos
  • guanacos Lama guanicoe
  • 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 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: 1, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 or SEQ ID NO: 51, including post-translational modifications thereof.
  • a heavy chain 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.
  • 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, SEQ ID NO:
  • 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 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: 3, SEQ ID NO: 18, SEQ ID NO:
  • a light chain 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, SEQ ID NO:
  • 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 AGE- KLH.
  • pAbs may be made in chickens, producing IgY molecules.
  • 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 locally, such as by intra-articular injection into a 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 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.
  • 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 a viral infection.
  • Viral infection may be determined by any suitable viral detection test, such as an antibody test, viral antigen detection test, viral culture or viral DNA or RNA detection test.
  • a subject may be considered to have received an effective antibody treatment if he or she demonstrates a reduction in viral infection symptoms between subsequent measurements or over time. Alternatively, the concentration and/or number of senescent cells may be measured over time. Administration of antibody and subsequent testing may be repeated until the desired therapeutic result is achieved.
  • 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, AG E-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 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-a nd-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 labile enterotoxin
  • silica particles 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.
  • NK natural killers
  • macrophages for example by macrophages
  • 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 hydroxyphosphate.
  • 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.
  • an immunogenic component for example squalene (MF-59) or mannide oleate (Incomplete Freund's Adjuvants)
  • MF-59 squalene
  • mannide oleate incomplete Freund's Adjuvants
  • 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 at. “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 sterilized
  • 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.
  • booster vaccinations may be desirable, over the course of 1 year to 20 years, including 2, 5,
  • 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. 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 a viral infection.
  • a subject may be considered to have received an effective vaccination if he or she demonstrates a reduction in viral symptoms between subsequent measurements or over time, or by 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.
  • the vaccine may contain multiple AGE antigens.
  • a subject may receive multiple vaccines, where each vaccine contains a different AGE antigen.
  • Any organism that is susceptible to viral infection such as mammals, 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. [106] A subject may be identified as in need of treatment based on a diagnosis with a viral infection, or with a disease caused by a viral infection.
  • viruses examples include herpesvirus, poxvirus, hepadnavirus, asfivirus, flavivirus, alphavirus, togavirus, coronavirus, hepatitis D, orthomyxovirus, paramyxovirus, rhabdovirus, bunyavirus, filovirus, human respiratory syncytial virus, retroviruses, adenoviruses, papilloma viruses, polyomavirus, Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), hepatitis B virus (HBV), hepatitis C virus (HCV), herpes simplex virus (HSV), human papilloma virus (HPV), Kaposi's sarcoma-associated herpesvirus (KSHV), human immunodeficiency virus (HIV), poliovirus, dengue virus and zika virus.
  • EBV Epstein-Barr virus
  • HCMV human cytomegalovirus
  • HBV hepatitis B
  • Rapidly replicating viruses are preferred viral infections for treatment.
  • examples of rapidly replicating viruses include influenza, such as influenza A virus subtype H5N1, coronaviruses, such as Middle East respiratory syndrome-related coronavirus (MERS-CoV) and severe acute respiratory syndrome-related coronavirus (SARS-CoV and SARS-CoV-2), and Ebola virus.
  • MERS-CoV Middle East respiratory syndrome-related coronavirus
  • SARS-CoV-2 severe acute respiratory syndrome-related coronavirus
  • Ebola virus Ebola virus.
  • SARS-CoV-2 is a preferred viral infection for treatment.
  • Subjects may also be identified as in need of treatment based on detection of advanced glycation end products in a sample obtained from the subject. Suitable samples include blood, skin, serum, saliva and urine. The diagnostic use of anti- AGE antibodies is discussed in more detail in International Patent Application Publication No. WO 2018/204679.
  • the Present Application includes 66 nucleotide and amino acid sequences in the Sequence Listing filed herewith. Variants of the nucleotide and amino acid sequences are possible. Known variants include substitutions, deletions and additions to the sequences shown in SEQ ID NO: 4, 16 and 20. In SEQ ID NO: 4, the arginine (Arg or R) residue at position 128 may optionally be omitted. In SEQ ID NO: 16, the alanine residue at position 123 may optionally be replaced with a serine residue, and/or the tyrosine residue at position 124 may optionally be replaced with a phenylalanine residue. SEQ ID NO: 20 may optionally include the same substitutions as SEQ ID NO: 16 at positions 123 and 124. In addition, SEQ ID NO: 20 may optionally contain one additional lysine residue after the terminal valine residue. [109] EXAMPLES
  • 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.
  • 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 immobilized 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.
  • 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.
  • Antibody purity was evaluated by capillary electrophoresis sodium-dodecyl sulfate (CE-SDS) analysis using LabChip® GXII, (PerkinElmer).
  • 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 1N HCI. 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 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.
  • 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 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 a/., “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:
  • Antibody humanness [144] 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.
  • 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 influenza.
  • 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 SARS-CoV.
  • 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 AG E-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 Ebola virus.
  • 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 SARS-CoV-2.
  • 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.
  • 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 10 6 cells/mL on the day of implant.
  • Tumors were initiated by subcutaneously implanting 1 x 10 5 4T1 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 mm 3 .
  • Tumor weight was approximated using the assumption that 1 mm 3 of tumor volume has a weight of 1 mg.
  • the four treatment groups are shown in Table 8 below:
  • 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
  • %TGI (1- MTVtreated/MTVcontrol) X 100.
  • %lnhibition (1-Mean Count of Focitreated/Mean Count of Focicontroi) x 100.
  • 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.
  • the %lnhibition was statistically significant (P ⁇ 0.01, ANOVA-Dunnett) for the 10 pg/g treatment group.
  • the ability of the carboxymethyl lysine antibody to treat cachexia trended towards significance (P > 0.05, ANOVA) based on a comparison of the organ weights of the lung and gastrocnemius between treatment groups and the control group.
  • the results indicate that administration of an anti-carboxymethyl lysine monoclonal antibody is able to reduce cancer metastases. This data provides additional evidence that in vivo administration of anti-AGE antibodies can provide therapeutic benefits safely and effectively.
  • Example 13 Treatment of a human subject with COVID-19 by administration of an anti-glycation end-product antibody (prophetic)
  • a human subject is diagnosed with COVID-19 due to infection with SARS- CoV-2.
  • the subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody).
  • anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the virus to obtain energy for replication.
  • the removal of AGE- modified cells deprives the virus of the energy needed for replication, resulting in reduction of the viral infection.
  • the subject recovers from COVID-19.
  • Example 14 treatment of a human subject with an intracellular bacterial infection (prophetic)
  • a human subject is diagnosed with an intracellular bacterial infection.
  • the subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody).
  • anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the bacteria to obtain energy for replication.
  • the removal of AGE-modified cells deprives the bacteria of the energy needed for replication, resulting in reduction of the infection.
  • Example 15 treatment of a human subject with an intracellular parasitic infection (prophetic)
  • a human subject is diagnosed with an intracellular parasite infection.
  • the subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody).
  • anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the parasite to obtain energy for replication.
  • the removal of AGE-modified cells deprives the parasite of the energy needed for replication, resulting in reduction of the infection.
  • Example 16 antibody binding to influenza virus infected cells
  • PRET Primary renal epithelial tubular cells were infected with 3 different viral concentrations of Influenza A H3N2/Wisconsin strain and incubated for approximately 24 hours. The 3 viral concentrations are referred to by their multiplicities of infection (MOI). After the incubation period, different antibody concentrations and antibody incubation periods were tested.
  • MOI multiplicities of infection
  • FIG. 2 shows the number of counts for an antibody concentration of 20 pg/mL and an antibody incubation period of 30 minutes.
  • FIG. 3 shows the number of counts for an antibody concentration of 5 pg/mL and an antibody incubation period of 60 minutes.
  • the red peaks correspond with an MOI of 1.0; yellow peaks correspond with an MOI of 0.1 ; cyan peaks correspond with an MOI of 0.01 ; and blue peaks correspond with uninfected cells.
  • the peak heights represent the number of events counted, in this case, the number of antibodies bound with the cells.
  • the shift of the peaks to the right indicates an increase in fluorescence intensity, and an increased intensity indicates a greater number of antibodies bound to the cells.
  • the antibody administered is a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody).
  • Tables 10 and 11 show the number of counts for each of the MOI peaks in FIG. 2 and FIG. 3, respectively.
  • the viral infection causes the cells to become more metabolically active, which increases the AGEs on the cell surface.
  • the MOI of 0.01 and 0.1 have the highest counts, as shown in FIG. 2 and FIG. 3.
  • the uninfected cells have lower counts than the MOI of 0.01 and 0.1 groups, which shows that the uninfected cells have a lower level of surface CML compared to the infected cells.

Abstract

A method of treating an infection that comprises administering to a subject a composition comprising an anti-AGE antibody. 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. A method of treating an infection that comprises immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.

Description

METHODS OF TREATING INFECTIONS
BACKGROUND
[01] Viruses are infectious agents that contain genetic material in the form of DNA or RNA within a protein coat known as a capsid. A complete virus particle of genetic material within a capsid is referred to as a virion. Some viruses have a viral envelope around the capsid. The viral envelope is typically composed of portions of cell membranes derived from a host cell and may also include viral glycoproteins. Examples of viruses that include a viral envelope include herpesvirus, poxvirus, hepadnavirus, asfivirus, flavivirus, alphavirus, togavirus, coronavirus, hepatitis D, orthomyxovirus, paramyxovirus, rhabdovirus, bunyavirus, filovirus and retroviruses.
[02] A viral infection occurs when a host organism is introduced to a pathogenic virus that replicates inside the cells of the organism. Viral replication is the process by which viruses take over or “hijack” the host cells to obtain energy and manufacture new viruses to facilitate the spread of the viral infection, both within the host and to new hosts.
[03] Rapidly replicating viruses are of particular concern because of their ability to quickly spread and infect new hosts. Widespread viral infections can progressively grow into outbreaks, epidemics and pandemics. Rapidly replicating viruses can also overwhelm the host organism, which may lead to organ damage or death. Examples of rapidly replicating viruses include influenza, such as influenza A virus subtype H5N1, coronaviruses, such as Middle East respiratory syndrome-related coronavirus (MERS-CoV) and severe acute respiratory syndrome-related coronavirus (SARS- CoV and SARS-CoV-2), and Ebola virus. These viruses have caused epidemics of the diseases known as the bird flu, MERS (Middle-East respiratory syndrome) and SARS (severe acute respiratory syndrome), and the pandemic of the disease known as COVID-19.
[04] There are no approved treatments to prevent or treat COVID-19, the disease that is caused by SARS-CoV-2. This virus has caused a pandemic that has led to approximately 3,000,000 confirmed infections and more than 200,000 deaths as of April 29, 2020 (“Coronavirus disease (COVID-19) Pandemic”, World Health Organization, available online at www.who.int/emergencies/diseases/novel- coronavirus-2019, accessed April 29, 2020). Unfortunately, this virus continues to spread globally and is projected to cause millions of deaths if no effective interventions are found and used. Vaccines and new drugs are being developed but their efficacies are still uncertain, and they will take at least a year or more to become available. An effective drug that is already known to be safe for human use would provide an ideal treatment.
[05] Pulmonary pathology in early-phase COVID-19 pneumonia has shown exudative and proliferative phases of acute lung injury (edema, inflammatory infiltrates, pneumocyte hyperplasia) prior to the development of any respiratory symptoms. In the later stage of disease, patients can develop acute respiratory distress syndrome (ARDS) and multi-organ failure. ARDS has taken center stage as the primary cause of death in the global COVID-19 crisis. The “cytokine storm” is emerging as a key mechanism leading to patient deterioration and death. Cytokine storm and ARDS has been associated with death from other viral infections, including SARS (SARS-COV), influenza viruses and the Spanish flu virus which caused the 1918 pandemic.
[06] Cytokine storm is associated with marked morbidity and mortality in patients presenting during the COVID-19 pandemic. The overwhelming progression to pulmonary failure is the hallmark vicious cycle overwhelming worldwide healthcare resources. Even a modest improvement in decreasing ventilator dependence, for example 5-10%, could dramatically alter outcomes in many healthcare settings.
[07] Some bacteria may accumulate inside cells, for example Mycobacterium tuberculosis and Pseudomonas aeruginosa. M. tuberculosis causes the formation of hard nodules or tubercles in the lungs, parasitizes macrophages by blocking the phagosome-lysosome fusion, a process called phagosome maturation arrest, and by replicating inside the phagosome (Vergne I, et al. Cell Biology of Mycobacterium tuberculosis Phagosome, Ann Rev Cell Dev Biol., Vol. 20, 367-94 (2004)). Similarly, P. aeruginosa colonizes the lungs of patients with cystic fibrosis and produces biofilms, alginates, and specific lipid A modifications, which allow the bacteria to escape immune response and cause severe chronic inflammation (Moskowitz SM, et al. The Role of Pseudomonas Lipopolysaccharide in Cystic Fibrosis Airway Infection, Subcell Biochem., Vol. 53, 241-53 (2010)). Production of biofilms by Haemophilus influenzae, Streptococcus pneumoniae, and other bacteria, has been linked to chronic otitis media in pediatric patients (Hall-Stoodley L, et al. Direct Detection of Bacterial Biofilms on the Middle-Ear Mucosa of Children With Chronic Otitis Media, JAMA, Vol. 256, No. 2, 202-11 (2006)).
[08] Some protozoan parasites present intracellular accumulation, for example
Plasmodium, Leishmania, Trypanosoma and Toxoplasma. Plasmodium, the agent causing malaria, replicates and accumulates inside erythrocytes, provoking cell rupture and dissemination of the agent, while the main sites of sequestration of the infected erythrocytes containing the trophozoites, schizonts and gametocytes of the parasite have been shown to be the lung, spleen, and adipose tissue, but also the brain, skin, bone marrow, and skeletal and cardiac muscle (Franke-Fayard B, etal. Sequestration and Tissue Accumulation of Human Malaria Parasites: Can We Learn Anything from Rodent Models of Malaria?, PLoS Pathogens, Vol. 6, No. 9, e1001032 (2010)). Similarly, Leishmania mexicana and Trypanosoma cruzi reside and proliferate inside macrophages (Zhang S et al. Delineation of Diverse Macrophage Activation Programs in Response to Intracellular Parasites and Cytokines, PLoS Negl Trop Dis, Vol. 4, No. 3: e648 (2010)).
[09] Many fungi are parasites on plants, animals (including humans), and other fungi. Fungus may invade tissue and can cause a disease. Some fungi may cause serious disease in humans. Fungi can attack eyes, nails, hair, and especially skin. One common fungal infection is Valley Fever, which is caused by Coccidioides immitis (CF). Valley Fever usually occurs due to inhalation of the arthroconidial spores of CF after soil disruption. Once inhaled, the spores enter the alveoli and enlarge in size to become spherules, and internal septations develop. Septations develop and form endospores within the spherule. The rupture of the spherules releases the endospores, which in turn repeat the cycle and spread the infection to adjacent tissues within the body.
[10] 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 b-galactosidase. Senescence begins with damage or stress (such as overstimulation by growth factors) of cells.
[11] Advanced glycation end-products (AGEs; also referred to as AGE-modified proteins or peptides, or glycation end-products) arise from a non-enzymatic reaction of sugars with protein side-chains (Ando, K. et ai, 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 and oxidative stress promote this post-translational modification of membrane proteins (Lindsey JB, etal., “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 inflammation, 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)).
[12] AGE-modified proteins are also a marker of senescent cells. This association between AGEs and senescence is well known in the art. See, for example, Gruber, L. (WO 2009/143411 , 26 Nov. 2009), Ando, K. etal. (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)), Ahmed, E.K. et al. (“Protein Modification and Replicative Senescence of WI-38 Human Embryonic Fibroblasts” Aging Cells, vol. 9, 252, 260 (2010)), Vlassara, H. etal. (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 end- products is associated with senescence and lack of function.
[13] 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 glyoxal. Glyoxal in turn reacts with proteins or lipids to generate advanced glycation end products. In the case of the protein component lysine, glyoxal reacts to form carboxymethyllysine, which is an AGE.
[14] 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).
[15] 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 hyperprol iterative state. However, there has been some ambiguity in this regard, as some tumors show overexpression of p16, while others 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. etal., p16lnk4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors, Oncogene, Vol. 30, 2087-2097 (2011)).
[16] 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. etal. “Interleukin-1 b 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-Ib, IL-5, IL-6, IL-8, IL-12, IL-23, CD2, CD3, CD20, CD22, CD52, CD80, CD86, C5 complement protein, BAFF, APRIL, IgE, a4b1 integrin and a4b7 integrin. Senescent cells also upregulate genes with roles in inflammation including IL-1 b, IL-8, ICAM1, TNFAP3, ESM1 and CCL2 (Burton, D.G.A. etal., “Microarray analysis of senescent vascular smooth muscle cells: a link to atherosclerosis and vascular calcification”, Experimental Gerontology, Vol. 44, No. 10, pp. 659-665 (October 2009)). 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.
[17] Senescent cells secrete reactive oxygen species (“ROS”) as part of the SASP. ROS are 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. etal., Cellular senescence and tumor suppressor gene p16, IntJ Cancer, Vol. 130, 1715-1725 (2012)).
[18] 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.
[19] Additional In vivo animal studies in mice found that removing senescent cells using senolytic agents treats aging-related disorders and atherosclerosis. Shortterm treatment with senolytic drugs in chronologically aged or progeroid mice alleviated several aging-related phenotypes (Zhu, Y. etal., “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. etal., “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.
[20] 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.
[21] 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
[22] In a first aspect, the invention is a method of treating an infection comprising administering to a subject a composition comprising an anti-AGE antibody.
[23] In a second aspect, the invention is a method of treating an infection 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.
[24] In a third aspect, the invention is a method of treating an infection comprising a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating the infection; followed by a second administering of the anti-AGE antibody.
[25] In a fourth aspect, the invention is use of an anti-AGE antibody for the manufacture of a medicament for treating an infection.
[26] In a fifth aspect, the invention is a composition comprising an anti-AGE antibody for use in treating an infection.
[27] In a sixth aspect, the invention is a composition for treating an infection 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.
[28] In a seventh aspect, the invention is a method of treating an infection comprising immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.
[29] In an eighth aspect, the invention is a method of treating an infection 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.
[30] In a ninth aspect, the invention is use of an AGE antigen for the manufacture of a medicament for treating an infection.
[31] In a tenth aspect, the invention is a composition comprising an AGE antigen for use in treating an infection.
[32] DEFINITIONS
[33] The term “peptide” means a molecule composed of 2-50 amino acids.
[34] The term “protein” means a molecule composed of more than 50 amino acids. [35] The terms “advanced glycation end-product”, “AGE”, “AGE-modified protein”, “AGE-modified 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)-1 H-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.
[36] 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.
[37] “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.
[38] 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 p10ink4a or expression of senescence-associated b-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.
[39] 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.
[40] 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, CA), 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.
[41] 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
[42] FIG. 1 is a graph of the response versus time in an antibody binding experiment.
[43] FIG. 2 is a graph of the number of events and the fluorescence intensity for antibodies bound to cells having various multiplicities of infection (MOI) of influenza virus.
[44] FIG. 3 is a graph of the number of events and the fluorescence intensity for antibodies bound to cells for various multiplicities of infection (MOI) of influenza virus. DETAILED DESCRIPTION
[45] Viruses must reprogram the host cell metabolism to increase the supply of nutrients, energy and metabolites that are necessary for replication. Viral control over host-cell metabolism involves upregulation of a carbon source, typically glucose or glutamine, and a redirection of these carbon supplies to metabolic pathways (Mayer, K.A. eta!., “Hijacking the supplies: metabolism as a novel facet of virus-host interaction”, Frontiers in Immunology, Vol. 10, Article 1533, 12 pages (2019)). Viruses obtain energy from the additional glucose through glycolysis (the enzymatic break down of glucose) and glycosylation (the enzymatic process that attaches glycansto proteins) (“Novel Coronavirus COVID-19”, Moleculin Biotech, available online at www.moleculin.com/covid-19/ (accessed April 28, 2020)). Enhanced glycolysis is also observed in cancer and oncogenic viruses and is known as aerobic glycolysis or the Warburg effect (Yu, L etal., “Oncogenic virus-induced aerobic glycolysis and tumorigenesis”, Journal of Cancer, Vol. 9, No. 20, pp. 3699-3706 (2018)).
[46] COVID-19 has been shown to rely on both glycolysis and glycosylation to fuel its growth. The characteristic spikes that surround coronaviruses such as SARS- CoV-2 are glycoproteins, which are formed by glycosylation. Multiple studies have shown that disrupting glycolysis and glycosylation is effective in stopping viruses like coronavirus (Moleculin Biotech). For example, the glucose decoy 2-deoxy-D- glucose (2-DG) has been shown to block glycolysis and completely prevent SARS- CoV-2 replication in human cells (Bojkova, D. etal., “SARS-CoV-2 infected host cell proteomics reveal potential therapy targets”, In Review Nature Research, available online atwww.researchsquare.com/article/rs-17218/v1, accessed April 29, 2020). These studies suggest that the altered metabolic pathways exhibited by virally- infected cells create novel therapeutic targets.
[47] Reactive oxygen species (ROS) are a natural byproduct of cellular metabolism. The increased cellular metabolism of infected host cells during viral replication results in a corresponding increase in reactive oxygen species. Respiratory viruses, such as influenza viruses and coronaviruses, show a marked increase in production of reactive oxygen species (Khomich, O.A. et al ., “Redox biology of respiratory viral infections”, Viruses, Vol. 10, No. 392, 27 pages (2018)). These reactive oxygen species cause oxidative stress/damage to cells as well as DNA damage, which has been shown to activate cellular senescence mechanisms in the respiratory virus human respiratory syncytial virus (HRSV) (Khomich, O.A. et a/.). The oxidative damage in turn leads to the formation of advanced glycation end- products (AGEs) through glycation, the non-enzymatic counterpart to glycosylation. Antibodies that bind to advanced glycation end-products (anti-AGE antibodies) have been shown to effectively treat age-related diseases such as sarcopenia (US 9,161 ,810) and metastatic cancer (WO 2017/143073) by binding and removing AGE- modified cells, such as senescent cells. Anti-AGE antibodies may be similarly used to bind cells that have been AGE-modified as a result of increased metabolic activity due to viral infection, such as highly glycolytic and glycoxidative cells.
[48] This enhanced glycolysis is also observed in bacterial, parasitic and fungal infections. Mycobacterial infections have been found to increase levels of AGEs (Rachman, H. et al., “Critical role of methylglyoxal and AGE in mycobacteria-induced macrophage apoptosis and activation”, PLOS One, issue 1, e29, pp. 1-8 (2006)).
The Warburg effect is observed in cells infected with intracellular bacteria, such as tuberculosis infections (Shi, L et al., “Biphasic dynamics of macrophage inmmunometabolism during Mycobacterium tuberculosis infection” mBio, Vol. 10,
No. 2, pp. 1-19 (2019) and Escroll, P. et al., “Metabolic reprogramming of host cells upon bacterial infection: Why shift to a Warburg-like metabolism?”, The FEBS Journal, Vol. 285, No. 12, pp. 2146-2160 (2018)). Bacterial infections have been found to turn on the production of ROS, which may delay the host response to the infection (Boncompain, G. et al., “Production of Reactive Oxygen Species Is Turned On and Rapidly Shut Down in Epithelial Cells Infected with Chlamydia trachomatis’’, Infection and Immunity, Vol. 78, No. 1 , pp. 80-87 (2010)). The enhanced glycolysis would also be expected to occur in parasite infections, as parasite reproduction inside cells requires energy (see Traore, K. et al., “Do advanced glycation end- products play a role in malaria susceptibility?”, Parasite, Vol. 23, No. 15, pp. 1-10 (2016)). Malarial parasites use aerobic glycolysis, and may produce elevated levels of carboxymethyllysine. (Sturm, et al. “Mitochondrial ATP synthase is dispensable in blood-stage Plasmodium berghei rodent malaria but essential in the mosquito phase”, PNAS, Vol. 112, No. 33, pp. 10216-10223 (2015)).
[49] Anti-AGE antibodies may also be used to bind AGE-modified proteins or peptides present on a viral envelope. Glycoproteins present in the viral envelope may be glycated due to the elevated level of reactive oxygen species and oxidative stress that occur during viral replication. The glycation of viral envelope glycoproteins forms AGEs, such as carboxymethyllysine, that will be recognized by anti-AGE antibodies. In addition, since the viral envelope often includes portions of the host cell membrane from which it was formed, enveloped viruses that replicate in glycated cells may retain AGE-modified proteins or peptides from the host cell membrane. Anti-AGE antibodies will bind to these formerly cell-surface AGE- modified proteins or peptides present on virions.
[50] The inflammatory response observed in viral infections also indicates that anti-AGE antibodies would be effective therapies against viral infections. The inflammatory response that is observed in viral infections is similarly observed with bacterial infections and parasitic infections. Interferons, particularly inflammatory cytokines, are produced during viral infections by the immune system (Eisenreich,
W. et al., “How viral and intracellular bacterial pathogens reprogram the metabolism of host cells to allow their intracellular replication", Frontiers in Cellular and Infection Microbiology, Vol. 9, Article 42, 33 pages (2019)). The cytokine storm is a systemic inflammatory response that is characterized by the release of Inflammatory cytokines. Senescent cells are known to secrete inflammatory factors and reactive oxygen species as part of the senescence-associated secretory phenotype (SASP), and the removal of these AGE-modified cells has been used to treat inflammation and auto-immune disorders (WO 2016/044252). The role of the cytokine storm observed in COVID-19 suggests that removal of AGE-modified cells would be similarly effective at reducing the inflammatory aspects of infections.
[51] The increased oxidative state resulting from the amplified metabolic activity during viral, bacterial, parasitic and fungal replication combined with the increased inflammatory environment from the cytokine storm indicates that AGE-modified cells are an appropriate therapeutic target for treating viral, bacterial, parasitic and fungal infections. The targeted removal of AGE-modified cells will reduce oxidative damage and reduce inflammation. The present invention uses enhanced clearance of cells expressing AGE-modified proteins or peptides (AGE-modified cells) to treat a viral infection. The present invention also includes enhanced clearance of cells expressing AGE-modified proteins or peptides (AGE-modified cells) to treat a bacterial, parasitic or fungal invention. This may be accomplished by administering anti-AGE antibodies to a subject. The anti-AGE antibodies may also bind to AGEs present on the viral envelope to remove virions and viruses or AGEs present on bacteria, parasites or fungi.
[52] 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 AGE-modified cells. The process of AGE-modified cell removal or destruction allows vaccination against AGE-modified proteins or peptides of a cell to be used to treat a viral infection, bacterial infection or parasitic infection. Vaccination against AGE-modified proteins or peptides also allows the immune system to target AGEs present on the viral envelope of virions and viruses, as well as bacteria and parasites.
[53] 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 etai). The antibody may bind to one or more AGE-modified proteins or peptides 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), camelized (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.
[54] 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 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), 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.
[55] 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 103, 7 x 103 or 6 x 103 (sec 1). 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 106, 8 x 106, 7 x 106, 6 x 106, 5 x 106, 4 x 106 or 3 x 106 (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.
[56] The anti-AGE antibody may destroy AGE-modified cells through antibody- dependent 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.
[57] 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.
[58] A toxin, such as pore-forming toxins (PFT) (Aroian R. et al., “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.
[59] 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.
[60] 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.
[61] Antibodies are Y-shaped proteins composed of two heavy chains and two light chains. The two arms of the Y shape form the fragment antigen-binding (Fab) region while the base or tail of the Y shape forms the fragment crystallizable (Fc) region of the antibody. Antigen binding occurs at the terminal portion of the fragment antigen-binding region (the tips of the arms of the Y shape) at a location referred to as the paratope, which is a set of complementarity determining regions (also known as CDRs or the hypervariable region). The complementarity determining regions vary among different antibodies and gives a given antibody its specificity for binding to a given antigen. The fragment crystallizable region of the antibody determines the result of antigen binding and may interact with the immune system, such as by triggering the complement cascade or initiating antibody-dependent cell-mediated cytotoxicity (ADCC). 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 shape being specific to one of the antigens; these are referred to as bi-specific antibodies.
[62] 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).
[63] The heavy chain of a humanized anti-AGE antibody 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 a humanized anti-AGE antibody 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 preparation of non-human anti-AGE antibodies.
[64] 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.
[65] 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.
[66] Particularly preferred anti-AGE antibodies may be obtained by humanizing murine monoclonal anti-AGE antibodies. Murine monoclonal anti-AGE antibodies have the heavy chain protein sequence shown in SEQ ID NO: 47 (the protein sequence of the variable domain is shown in SEQ ID NO: 52) and the light chain protein sequence shown in SEQ ID NO: 57 (the protein sequence of the variable domain is shown in SEQ ID NO: 62). A preferred humanized heavy chain may have the protein sequence shown in SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 or SEQ ID NO: 51 (the protein sequences of the variable domains of the humanized heavy chains are shown in SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55 and SEQ ID NO: 56, respectively). A preferred humanized light chain may have the protein sequence shown in SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60 or SEQ ID NO: 61 (the protein sequences of the variable domains of the humanized light chains are shown in SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65 and SEQ ID NO: 66, respectively). Preferably, a humanized anti-AGE monoclonal antibody is composed a heavy chain having a protein sequence selected from the group consisting of SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51 and a light chain having a protein sequence selected from the group consisting of SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60 and SEQ ID NO: 61. Humanized monoclonal anti-AGE antibodies composed of these protein sequences may have better binding and/or improved activation of the immune system, resulting in greater efficacy.
[67] 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).
[68] 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).
[69] 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.
[70] 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).
[71] An anti-AGE antibody or a variant thereof may include a heavy chain 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: 1, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 or SEQ ID NO: 51, including post-translational modifications thereof. A heavy chain 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.
[72] 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, SEQ ID NO:
20, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 41, SEQ ID NO:
42, SEQ ID NO: 43, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO:
55, or SEQ ID NO: 56, 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.
[73] An anti-AGE antibody or a variant thereof may include a light chain 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: 3, SEQ ID NO: 18, SEQ ID NO:
19, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 39, SEQ ID NO: 57, SEQ ID NO:
58, SEQ ID NO: 59, SEQ ID NO: 60 or SEQ ID NO: 61, including post-translational modifications thereof. A light chain 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.
[74] 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, SEQ ID NO:
21, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65 or SEQ ID NO: 66, 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.
[75] 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).
[76] The antibody may have or may include constant regions which permit destruction of targeted cells by a subject’s immune system.
[77] Mixtures of antibodies that bind to more than one type AGE of AGE-modified proteins may also be used.
[78] 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.
[79] 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.
[80] 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- -N-acetyl hexokinase, AGE-apo E, AGE-red cell membrane protein, AGE-aldose reductase, AGE-ferritin, AGE-red cell spectrin, AGE-alcohol dehydrogenase, AG E-haptoglobin, AGE-tubulin, AGE-thyroid hormone, AGE-fibrinogen, AGE- 2-microglobulin, AGE- sorbitol dehydrogenase, AGE-cu-antitrypsin, AGE-carbonate dehydratase, AGE- RNAse, AGE-hexokinase, AGE-apo C-l, 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.
[81] 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.
[82] 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.
[83] The antibodies may be administered by injection, such as by intravenous injection or locally, such as by intra-articular injection into a 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.
[84] 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.
[85] 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.
[86] A subject that receives administration of an anti-AGE antibody may be tested to determine if the administration has been effective to treat a viral infection. Viral infection may be determined by any suitable viral detection test, such as an antibody test, viral antigen detection test, viral culture or viral DNA or RNA detection test. A subject may be considered to have received an effective antibody treatment if he or she demonstrates a reduction in viral infection symptoms between subsequent measurements or over time. Alternatively, the concentration and/or number of senescent cells may be measured over time. Administration of antibody and subsequent testing may be repeated until the desired therapeutic result is achieved.
[87] 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.
[88] 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, AG E-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 hexokinase, 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-hexokinase, AGE-apo C-l, 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.
[89] 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 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-a nd-reagents) .
[90] 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.
[91] 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.
[92] 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.
[93] 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.
[94] 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 hydroxyphosphate. 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. [95] 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.
[96] 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.
[97] 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 at. “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.
[98] 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.
[99] 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/B/excipient-table-2.pdf and Vogel, F. R. etal., “A compendium of vaccine adjuvants and excipients”, Pharmaceutical Biotechnology, Vol. 6, pp. 141-228 (1995).
[100] 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.
[101] 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. [102] 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.
[103] 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. 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 a viral infection. A subject may be considered to have received an effective vaccination if he or she demonstrates a reduction in viral symptoms between subsequent measurements or over time, or by measuring the concentration and/or number of senescent cells. Vaccination and subsequent testing may be repeated until the desired therapeutic result is achieved.
[104] 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.
[105] Any organism that is susceptible to viral infection, such as mammals, 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. [106] A subject may be identified as in need of treatment based on a diagnosis with a viral infection, or with a disease caused by a viral infection. Examples of viruses that may be treated include herpesvirus, poxvirus, hepadnavirus, asfivirus, flavivirus, alphavirus, togavirus, coronavirus, hepatitis D, orthomyxovirus, paramyxovirus, rhabdovirus, bunyavirus, filovirus, human respiratory syncytial virus, retroviruses, adenoviruses, papilloma viruses, polyomavirus, Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), hepatitis B virus (HBV), hepatitis C virus (HCV), herpes simplex virus (HSV), human papilloma virus (HPV), Kaposi's sarcoma-associated herpesvirus (KSHV), human immunodeficiency virus (HIV), poliovirus, dengue virus and zika virus. Rapidly replicating viruses are preferred viral infections for treatment. Examples of rapidly replicating viruses include influenza, such as influenza A virus subtype H5N1, coronaviruses, such as Middle East respiratory syndrome-related coronavirus (MERS-CoV) and severe acute respiratory syndrome-related coronavirus (SARS-CoV and SARS-CoV-2), and Ebola virus. SARS-CoV-2 is a preferred viral infection for treatment.
[107] Subjects may also be identified as in need of treatment based on detection of advanced glycation end products in a sample obtained from the subject. Suitable samples include blood, skin, serum, saliva and urine. The diagnostic use of anti- AGE antibodies is discussed in more detail in International Patent Application Publication No. WO 2018/204679.
[108] The Present Application includes 66 nucleotide and amino acid sequences in the Sequence Listing filed herewith. Variants of the nucleotide and amino acid sequences are possible. Known variants include substitutions, deletions and additions to the sequences shown in SEQ ID NO: 4, 16 and 20. In SEQ ID NO: 4, the arginine (Arg or R) residue at position 128 may optionally be omitted. In SEQ ID NO: 16, the alanine residue at position 123 may optionally be replaced with a serine residue, and/or the tyrosine residue at position 124 may optionally be replaced with a phenylalanine residue. SEQ ID NO: 20 may optionally include the same substitutions as SEQ ID NO: 16 at positions 123 and 124. In addition, SEQ ID NO: 20 may optionally contain one additional lysine residue after the terminal valine residue. [109] EXAMPLES
[110] Example 1 : In vivo study of the administration of anti-glycation end-product antibody
[111] 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.
[112] 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.
[113] Table 1 : The different groups of animals used in the study
Figure imgf000038_0001
Not Applicable, Pre : Subset of animals euthanized prior to treatment start for collection of adipose tissue.
[114] p-j0iNK4a 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 -DDei. [115] Table 2: pi6INK4a mRNA quantified in adipose tissue
Figure imgf000039_0001
[116] 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.
[117] 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.
[118] These results indicate the antibody administration resulted in the killing of senescent cells. [119] 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.
[120] Table 3: Effect of antibody administration on mass of the gastrocnemius muscle
Summary Absolute weight of Weight relative to body mass of
Group Information Gastrocnemius Muscle (g) Gastrocnemius 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
[121] 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. This data provides evidence that in vivo administration of anti-AGE antibodies can provide therapeutic benefits safely and effectively. [122] Example 2: Affinity and kinetics of test antibody
[123] 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 immobilized 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.
[124] Table 4: Experimental set-up of affinity and kinetics analysis
Figure imgf000041_0001
[125] 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; /¾ ( 1/s) = 6.781 x 10-3; KD (M) = 3.651 x 10-®; 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. [126] Example 3: Construction and production of murine anti-AGE lgG2b antibody and chimeric anti-AGE lgG1 antibody
[127] 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.
[128] 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.
[129] 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:
[130] Table 5: Yields and aliquots
Figure imgf000042_0001
Figure imgf000043_0001
[131] Antibody purity was evaluated by capillary electrophoresis sodium-dodecyl sulfate (CE-SDS) analysis using LabChip® GXII, (PerkinElmer).
[132] Example 4: Binding of murine (parental) and chimeric anti-AGE antibodies
[133] 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.
[134] 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 1N HCI. A second direct binding ELISA was performed with only CML coating. The absorbance at OD450 was read using a microplate reader.
[135] 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.
[136] Plate map of CML and CML-KLH ELISA:
Cone.
(pg/mL) 1 2 3 4 5 6 7
Figure imgf000044_0001
[137] 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.
[138] Plate map of CML-only ELISA:
Cone.
(pg/mL) 1 2 3 4 5 6 7
Figure imgf000044_0002
Figure imgf000045_0001
[139] 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.
[140] Example 5: Humanized antibodies
[141] 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.
[142] 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 a/., “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:
[143] Table 6: Antibody humanness
Figure imgf000046_0001
[144] 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.
[145] 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).
[146] 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:
[147] Table 7: Antibody titers
Figure imgf000047_0001
Figure imgf000048_0001
[148] The binding of the humanized antibodies may be evaluated, for example, by dose-dependent binding ELISA or cell-based binding assay.
[149] Example 6 (Prophetic): An AGE-RNAse containing vaccine in a human subject.
[150] 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 influenza.
[151] Example 7 (Prophetic): Injection regimen for an AGE-RNAse containing vaccine in a human subject.
[152] The same vaccine as described in Example 6 is injected intra-articularly into a subject with SARS-CoV. 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.
[153] Example 8 (Prophetic): An AG E-hemoglobin containing vaccine in a human subject.
[154] 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.
[155] Example 9 (Prophetic): An AGE-human serum albumin containing vaccine in a human subject.
[156] 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.
[157] Example 10: Carboxymethyllysine-modified protein vaccine for a human subject (prophetic)
[158] 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 Ebola virus.
[159] Example 11 : Carboxyethyllysine-modified peptide vaccine for a human subject (prophetic)
[160] 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 SARS-CoV-2. [161] Example 12: In vivo study of the administration of a carboxymethyl lysine monoclonal antibody
[162] 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.
[163] 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.
[164] 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:
[165] Table 8: Treatment groups
Figure imgf000050_0001
Figure imgf000051_0001
li 66] 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.
[167] 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.
[168] 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).
[169] 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
-so the control group. %TGI may be calculated according to the formula: %TGI = (1- MTVtreated/MTVcontrol) X 100.
[170] 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.
[171] 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.
[172] 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.
[173] 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.
[174] The results of the study are shown below in Table 9: [175] Table 9: Results
Figure imgf000053_0001
[176] 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 trended towards significance (P > 0.05, Mann-Whitney) for the 5 pg/g (Group 2) or 10 pg/g treatment group (Group 3). The %lnhibition trended towards significance (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. The ability of the carboxymethyl lysine antibody to treat cachexia trended towards significance (P > 0.05, ANOVA) based on a comparison of the organ weights of the lung and gastrocnemius between treatment groups and the control group. The results indicate that administration of an anti-carboxymethyl lysine monoclonal antibody is able to reduce cancer metastases. This data provides additional evidence that in vivo administration of anti-AGE antibodies can provide therapeutic benefits safely and effectively.
[177] Example 13: Treatment of a human subject with COVID-19 by administration of an anti-glycation end-product antibody (prophetic)
[178] A human subject is diagnosed with COVID-19 due to infection with SARS- CoV-2. The subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody). The anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the virus to obtain energy for replication. The removal of AGE- modified cells deprives the virus of the energy needed for replication, resulting in reduction of the viral infection. The subject recovers from COVID-19.
[179] Example 14: treatment of a human subject with an intracellular bacterial infection (prophetic)
[180] A human subject is diagnosed with an intracellular bacterial infection. The subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody). The anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the bacteria to obtain energy for replication. The removal of AGE-modified cells deprives the bacteria of the energy needed for replication, resulting in reduction of the infection.
[181] Example 15: treatment of a human subject with an intracellular parasitic infection (prophetic)
[182] A human subject is diagnosed with an intracellular parasite infection. The subject is administered a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody). The anti-CML antibody binds and destroys AGE-modified cells, interfering with the metabolic process used by the parasite to obtain energy for replication. The removal of AGE-modified cells deprives the parasite of the energy needed for replication, resulting in reduction of the infection.
[183] Example 16: antibody binding to influenza virus infected cells
[184] Primary renal epithelial tubular (PRET) cells were infected with 3 different viral concentrations of Influenza A H3N2/Wisconsin strain and incubated for approximately 24 hours. The 3 viral concentrations are referred to by their multiplicities of infection (MOI). After the incubation period, different antibody concentrations and antibody incubation periods were tested.
[185] FIG. 2 shows the number of counts for an antibody concentration of 20 pg/mL and an antibody incubation period of 30 minutes. FIG. 3 shows the number of counts for an antibody concentration of 5 pg/mL and an antibody incubation period of 60 minutes. The red peaks correspond with an MOI of 1.0; yellow peaks correspond with an MOI of 0.1 ; cyan peaks correspond with an MOI of 0.01 ; and blue peaks correspond with uninfected cells. The peak heights represent the number of events counted, in this case, the number of antibodies bound with the cells. The shift of the peaks to the right indicates an increase in fluorescence intensity, and an increased intensity indicates a greater number of antibodies bound to the cells. The antibody administered is a humanized anti-glycation end-product antibody raised against carboxymethyl lysine (anti-CML antibody). Tables 10 and 11 show the number of counts for each of the MOI peaks in FIG. 2 and FIG. 3, respectively.
[186] Table 10: 20 pg/mL of antibody and an incubation period of 30 minutes
Figure imgf000055_0001
[187] Table 11 : 5 pg/mL of antibody and an incubation period of 60 minutes
Figure imgf000055_0002
[188] The viral infection causes the cells to become more metabolically active, which increases the AGEs on the cell surface. The MOI of 0.01 and 0.1 have the highest counts, as shown in FIG. 2 and FIG. 3. The uninfected cells have lower counts than the MOI of 0.01 and 0.1 groups, which shows that the uninfected cells have a lower level of surface CML compared to the infected cells. These results indicate that viral infection increases the presence of surface CML on cells. At the highest MOI of 1.0, the count is lower due to cytopathic events caused by the virus or the loss of cells during washing cycles. As fewer cells survived in the MOI of 1.0 group, the resulting count is lower.
[189] REFERENCES
[190] 1. Eisenreich, W. et al., “How viral and intracellular bacterial pathogens reprogram the metabolism of host cells to allow their intracellular replication”, Frontiers in Cellular and Infection Microbiology, Vol. 9, Article 42, 33 pages (2019).
[191] 2. Mayer, K.A. et al., “Hijacking the supplies: metabolism as a novel facet of virus-host interaction”, Frontiers in Immunology, Vol. 10, Article 1533, 12 pages (2019).
[192] 3. “Novel Coronavirus COVID-19”, Moleculin Biotech, available online at www.moleculin.com/covid-19/ (accessed April 28, 2020).
[193] 4. “Coronavirus disease (COVID-19) Pandemic”, World Health
Organization, available online at www.who.int/emergencies/diseases/novel- coronavirus-2019, accessed April 29, 2020.
[194] 5. Bojkova, D. et al., “SARS-CoV-2 infected host cell proteomics reveal potential therapy targets", In Review Nature Research, available online at www.researchsquare.com/article/rs-17218/v1, accessed April 29, 2020).
[195] 6. Khomich, O.A. et al., “Redox biology of respiratory viral infections”,
Viruses, Vol. 10, No. 392, 27 pages (2018).
[196] 7. Yu, L et al., “Oncogenic virus-induced aerobic glycolysis and tumorigenesis”, Journal of Cancer, Vol. 9, No. 20, pp. 3699-3706 (2018).
[197] 8. Sanchez, E.L. et al., “Viral activation of cellular metabolism”, Virology,
Vol. 479-480, pp. 609-618 (2015).
[198] 9. Shi, L et al., “Biphasic dynamics of macrophage inmmunometabolism during Mycobacterium tuberculosis infection” mBio, Vol. 10, No. 2, pp. 1-19 (2019).
[199] 10. Rachman, H. et al., “Critical role of methylglyoxal and AGE in mycobacteria-induced macrophage apoptosis and activation”, PLOS One, issue 1 , e29, pp. 1-8 (2006). [200] 11. Traore, K. et al., “Do advanced glycation end-products play a role in malaria susceptibility?”, Parasite, Vol. 23, No. 15, pp. 1-10 (2016).
[201] 12. Escroll, P. et al., “Metabolic reprogramming of host cells upon bacterial infection: Why shift to a Warburg-like metabolism?”, The FEBS Journal, Vol. 285,
No. 12, pp. 2146-2160 (2018).
[202] 13. Boncompain, G. et al., “Production of Reactive Oxygen Species Is Turned On and Rapidly Shut Down in Epithelial Cells Infected with Chlamydia trachomatis ”, Infection and Immunity, Vol. 78, No. 1, pp. 80-87 (2010).
[203] 14. Vergne I, et al. Cell Biology of Mycobacterium tuberculosis Phagosome, Ann Rev Cell Dev Biol., Vol. 20, 367-94 (2004).
[204] 15. Moskowitz SM, et al. The Role of Pseudomonas Lipopolysaccharide in Cystic Fibrosis Airway Infection, Subcell Biochem., Vol. 53, 241-53 (2010).
[205] 16. Hall-Stoodley L, et al. Direct Detection of Bacterial Biofilms on the Middle-Ear Mucosa of Children With Chronic Otitis Media, JAMA, Vol. 256, No. 2, 202-11 (2006).
[206] 17. Franke-Fayard B, et al. Sequestration and Tissue Accumulation of Human Malaria Parasites: Can We Learn Anything from Rodent Models of Malaria?, PLoS Pathogens, Vol. 6, No. 9, e1001032 (2010).
[207] 18. Zhang S et al. Delineation of Diverse Macrophage Activation Programs in Response to Intracellular Parasites and Cytokines, PLoS Negl Trap Dis, Vol. 4, No. 3: e648 (2010).
[208] Sturm, et al. “Mitochondrial ATP synthase is dispensable in blood-stage Plasmodium berghei rodent malaria but essential in the mosquito phase”, PNAS,
Vol. 112, No. 33, pp. 10216-10223 (2015).

Claims

WHAT IS CLAIMED IS:
1. A method of treating an infection, comprising administering to a subject a composition comprising an anti-AGE antibody.
2. A method of treating an infection, 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 an infection, comprising: a first administering of an anti-AGE antibody; followed by testing the subject for effectiveness of the first administration at treating the infection; followed by a second administering of the anti-AGE antibody.
4. A composition for treating an infection, 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 an infection, comprising immunizing a subject in need thereof against AGE-modified proteins or peptides of a cell.
6. A method of treating a subject with an infection, 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 or composition of any of the preceding claims, wherein the infection is a viral infection.
8. The method or composition of any of the preceding claims, wherein the infection is a bacterial infection.
9. The method or composition of any of the preceding claims, wherein the infection is a parasitic infection.
10. The method or composition of any of the preceding claims, wherein the composition further comprises a pharmaceutically acceptable carrier.
11. The method or composition of any of the preceding claims, wherein the subject is selected from the group consisting of humans, goats, sheep, cows, horses, dogs and cats.
12. The method 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.
13. The method or composition of any of the preceding claims, wherein the anti-AGE antibody is administered intravenously.
14. The method or composition of any of the preceding claims, wherein the anti-AGE antibody is administered locally.
15. The method 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, carboxyethy I lysine and pentosidine.
16. The method or composition of any of the preceding claims, wherein the first anti-AGE antibody and the second anti-AGE antibody each independently 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.
17. The method or composition of any of the preceding claims, wherein the composition is in unit dosage form.
18. The method or composition of any of the preceding claims, wherein the composition is in multidosage form.
19. The method or composition of any of the preceding claims, wherein the composition is sterile.
20. The method or composition of any of the preceding claims, wherein the immunizing comprises administering a vaccine comprising an AGE antigen.
21. The method 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.
22. The method 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, AG E-antithrombin III, AGE-calmodulin, AGE-insulin, AGE-ceruloplasmin, AGE- collagen, AGE-cathepsin B, AGE-crystallin, AGE-plasminogen activator, AGE- endothelial plasma membrane protein, AGE-aldehyde reductase, AG E-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 hexokinase, 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- 2-microglobulin, AGE-sorbitol dehydrogenase, AGE-ai-antitrypsin, AGE-carbonate dehydratase, AGE-hexokinase, AGE-apo C-l, AGE-KLH and mixtures thereof.
23. The method or composition of any of the preceding claims, further comprising testing the patient to determine if the viral infection has been ameliorated, and repeating the immunizing, if necessary.
24. The method or composition of any of the preceding claims, further comprising testing the patient to determine if the bacterial infection has been ameliorated, and repeating the immunizing, if necessary.
25. The method or composition of any of the preceding claims, further comprising testing the patient to determine if the parasitic infection has been ameliorated, and repeating the immunizing, if necessary.
26. The method or composition of any of the previous claims, wherein the antibody comprises a first complementary determining region comprising the amino acid sequence of SEQ ID NO: 23, a second complementary determining region comprising the amino acid sequence of SEQ ID NO: 24, a third complementary determining region comprising the amino acid sequence of SEQ ID NO: 25, a fourth complementary determining region comprising the amino acid sequence of SEQ ID NO: 26, a fifth complementary determining region comprising the amino acid sequence of SEQ ID NO: 27, and a sixth complementary determining region comprising the amino acid sequence of SEQ ID NO: 28.
27. The method or composition of any of the previous claims, wherein the antibody comprises a first complementary determining region comprising the amino acid sequence of SEQ ID NO: 41, a second complementary determining region comprising the amino acid sequence of SEQ ID NO: 42, a third complementary determining region comprising the amino acid sequence of SEQ ID NO: 43, a fourth complementary determining region comprising the amino acid sequence of SEQ ID NO: 44, a fifth complementary determining region comprising the amino acid sequence of SEQ ID NO: 45, and a sixth complementary determining region comprising the amino acid sequence of SEQ ID NO: 46.
28. The method 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 , SEQ ID NO: 33, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, and SEQ ID NO: 51 , 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, SEQ ID NO: 39, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, and SEQ ID NO: 61.
29. The method or composition of any of the previous claims, wherein the antibody comprises a constant region from a species selected from the group consisting of humans, goats, sheep, pigs, cows, horses, camels, alpacas, dogs and cats.
30. The method or composition of any of the preceding claims, wherein the antibody is a humanized antibody.
31. The method or composition of any of the preceding claims, wherein the antibody is monoclonal.
32. The method or composition of any of the preceding claims, wherein the antibody is substantially non-immunogenic to humans.
33. The method or composition of any of the preceding claims, wherein the antibody has a rate of dissociation (kd) of at most 9 x 103 sec-1.
34. The method or composition of any of the preceding claims, wherein the antibody is conjugated to an agent that causes the destruction of AGE-modified cells.
35. The method 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.
36. The method 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.
37. The method or composition of any of the preceding claims, wherein the AGE antigen comprises carboxymethyl lysine conjugated with keyhole limpet hemocyanin (CML-KLH).
38. The method or composition of any of the preceding claims, wherein the viral infection comprises at least one viral infection selected from the group consisting of influenza, influenza A virus subtype H5N1 , coronaviruses, Middle East respiratory syndrome-related coronavirus (MERS-CoV), severe acute respiratory syndrome-related coronavirus (SARS-CoV), severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2), and Ebola virus.
39. The method or composition of any of the preceding claims, wherein the viral infection comprises severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2).
40. The method or composition of any of the preceding claims, wherein the viral infection is caused by at least one virus selected from the group consisting of herpesvirus, poxvirus, hepadnavirus, asfivirus, flavivirus, alphavirus, togavirus, coronavirus, hepatitis D, orthomyxovirus, paramyxovirus, rhabdovirus, bunyavirus, filovirus, human respiratory syncytial virus, retroviruses, adenoviruses, papilloma viruses, polyomavirus, Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), hepatitis B virus (HBV), hepatitis C virus (HCV), herpes simplex virus (HSV), human papilloma virus (HPV), Kaposi's sarcoma-associated herpesvirus (KSHV), human immunodeficiency virus (HIV) and poliovirus.
41 . The method or composition of any of the preceding claims, wherein the anti-AGE antibody binds an AGE-modified protein or peptide on a viral envelope.
42. The method or composition of any of the preceding claims, wherein the bacterial infection is caused by at least one bacterial species selected from the group consisting of: Chlamydia trachomatis, Mycobacterium tuberculosis and Pseudomonas aeruginosa.
43. The method or composition of any of the preceding claims, wherein the parasitic infection is caused by at least one parasite selected from the group consisting of: Plasmodium, Leishmania, Trypanosoma and Toxoplasma.
44. The method or composition of any of the preceding claims, wherein the infection is a fungal infection.
PCT/US2021/030184 2020-05-01 2021-04-30 Methods of treating infections WO2021222758A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
IL297799A IL297799A (en) 2020-05-01 2021-04-30 Methods of treating infections
CN202180047404.4A CN115996954A (en) 2020-05-01 2021-04-30 Methods of treating infections
CA3177449A CA3177449A1 (en) 2020-05-01 2021-04-30 Methods of treating infections
JP2022566616A JP2023524098A (en) 2020-05-01 2021-04-30 How to treat infections
AU2021264007A AU2021264007A1 (en) 2020-05-01 2021-04-30 Methods of treating infections
US17/922,264 US20230181730A1 (en) 2020-05-01 2021-04-30 Methods of treating infections
EP21731330.3A EP4143238A1 (en) 2020-05-01 2021-04-30 Methods of treating infections

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063019139P 2020-05-01 2020-05-01
US63/019,139 2020-05-01
US202063109757P 2020-11-04 2020-11-04
US63/109,757 2020-11-04

Publications (1)

Publication Number Publication Date
WO2021222758A1 true WO2021222758A1 (en) 2021-11-04

Family

ID=76355566

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/030184 WO2021222758A1 (en) 2020-05-01 2021-04-30 Methods of treating infections

Country Status (8)

Country Link
US (1) US20230181730A1 (en)
EP (1) EP4143238A1 (en)
JP (1) JP2023524098A (en)
CN (1) CN115996954A (en)
AU (1) AU2021264007A1 (en)
CA (1) CA3177449A1 (en)
IL (1) IL297799A (en)
WO (1) WO2021222758A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11213585B2 (en) 2016-06-23 2022-01-04 Siwa Corporation Vaccines for use in treating various diseases and disorders
US11261241B2 (en) 2008-05-23 2022-03-01 Siwa Corporation Methods, compositions and apparatuses for facilitating regeneration
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) 2017-04-14 2024-04-16 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders

Citations (13)

* 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
WO2017143073A1 (en) 2016-02-19 2017-08-24 Siwa Corporation Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (age)
WO2017181116A1 (en) * 2016-04-15 2017-10-19 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
WO2018204679A1 (en) 2017-05-04 2018-11-08 Siwa Corporation Diagnostic advanced glycation end-product antibodies
WO2020023532A1 (en) * 2018-07-23 2020-01-30 Siwa Corporation Methods and compositions for treating chronic effects of radiation and chemical exposure

Patent Citations (14)

* 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
US9161810B2 (en) 2008-05-23 2015-10-20 Siwa Corporation Methods, compositions and apparatuses for facilitating regeneration
WO2016044252A2 (en) 2014-09-19 2016-03-24 Siwa Corporation Anti-age antibodies for treating inflammation and auto-immune disorders
WO2017143073A1 (en) 2016-02-19 2017-08-24 Siwa Corporation Method and composition for treating cancer, killing metastatic cancer cells and preventing cancer metastasis using antibody to advanced glycation end products (age)
WO2017181116A1 (en) * 2016-04-15 2017-10-19 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
WO2018204679A1 (en) 2017-05-04 2018-11-08 Siwa Corporation Diagnostic advanced glycation end-product antibodies
WO2020023532A1 (en) * 2018-07-23 2020-01-30 Siwa Corporation Methods and compositions for treating chronic effects of radiation and chemical exposure

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. AJ245184
"Novel Coronavirus COVID-19", 28 April 2020, MOLECULIN BIOTECH
AHMED, E.K. ET AL.: "Protein Modification and Replicative Senescence of WI-38 Human Embryonic Fibroblasts", AGING CELLS, vol. 9, 2010, pages 252,260
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
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
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
BOJKOVA, D. ET AL.: "SARS-CoV-2 infected host cell proteomics reveal potential therapy targets", REVIEW NATURE RESEARCH, 29 April 2020 (2020-04-29)
BONCOMPAIN, G. ET AL.: "Production of Reactive Oxygen Species Is Turned On and Rapidly Shut Down in Epithelial Cells Infected with Chlamydia trachomatis", INFECTION AND IMMUNITY, vol. 78, no. 1, 2010, pages 80 - 87
BURTON, D.G.A. ET AL.: "Microarray analysis of senescent vascular smooth muscle cells: a link to atherosclerosis and vascular calcification", EXPERIMENTAL GERONTOLOGY, vol. 44, no. 10, October 2009 (2009-10-01), pages 659 - 665, XP026640753, DOI: 10.1016/j.exger.2009.07.004
EISENREICH, W. ET AL.: "How viral and intracellular bacterial pathogens reprogram the metabolism of host cells to allow their intracellular replication", FRONTIERS IN CELLULAR AND INFECTION MICROBIOLOGY, vol. 9, 2019
ESCROLL, P. ET AL.: "Metabolic reprogramming of host cells upon bacterial infection: Why shift to a Warburg-like metabolism?", THE FEBS JOURNAL, vol. 285, no. 12, 2018, pages 2146 - 2160
FARBOUD B ET AL: "Development of a Polyclonal Antibody with Broad Epitope Specificity for Advanced Glycation Endproducts and Localization of these Epitopes in Bruch's Membrane of the Aging Eye", MOLECULAR VISION, vol. 5, 14 July 1999 (1999-07-14), pages 11, XP055829985, Retrieved from the Internet <URL:<http://www.molvis.org/molvis/v5/p11>> *
FERRACCIOLI, G. ET AL.: "Interleukin-1 β 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., vol. 16, no. 11-12, November 2010 (2010-11-01), pages 552 - 557
FRANKE-FAYARD B ET AL.: "Sequestration and Tissue Accumulation of Human Malaria Parasites: Can We Learn Anything from Rodent Models of Malaria?", PLOS PATHOGENS, vol. 6, no. 9, 2010, pages e1001032
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
GAO, S. H.: "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
HALL-STOODLEY L ET AL.: "Direct Detection of Bacterial Biofilms on the Middle-Ear Mucosa of Children With Chronic Otitis Media", JAMA, vol. 256, no. 2, 2006, pages 202 - 11
IKEDA K ET AL: "Immunochemical approaches to AGE-structures: characterization of anti-AGE antibodies", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 215, no. 1-2, 1 June 1998 (1998-06-01), pages 95 - 104, XP004146496, ISSN: 0022-1759, DOI: 10.1016/S0022-1759(98)00064-7 *
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
KHOMICH, O.A. ET AL.: "Redox biology of respiratory viral infections", VIRUSES, vol. 10, no. 392, 2018
LINDSEY JB 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
MAYER, K.A. ET AL.: "Hijacking the supplies: metabolism as a novel facet of virus-host interaction", FRONTIERS IN IMMUNOLOGY, vol. 10, 2019
MOSKOWITZ SM ET AL.: "The Role of Pseudomonas Lipopolysaccharide in Cystic Fibrosis Airway Infection", SUBCELL BIOCHEM., vol. 53, 2010, pages 241 - 53
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
RACHMAN, H. ET AL.: "Critical role of methylglyoxal and AGE in mycobacteria-induced macrophage apoptosis and activation", PLOS ONE, 2006, pages 1 - 8
RAYESS, H. ET AL.: "Cellular senescence and tumor suppressor gene p16", INT J CANCER, vol. 130, 2012, pages 1715 - 1725, XP055282399, DOI: 10.1002/ijc.27316
ROMAGOSA, C. ET AL.: "p161nk4a 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
SANCHEZ, E.L. ET AL.: "Viral activation of cellular metabolism", VIROLOGY, vol. 479-480, 2015, pages 609 - 618
SHI, L ET AL.: "Biphasic dynamics of macrophage inmmunometabolism during Mycobacterium tuberculosis infection", MBIO, vol. 10, no. 2, 2019, pages 1 - 19
SON SEUNGHWAN ET AL: "Advanced glycation end products impair NLRP3 inflammasome-mediated innate immune responses in macrophages", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 292, no. 50, 1 December 2017 (2017-12-01), US, pages 20437 - 20448, XP055829811, ISSN: 0021-9258, DOI: 10.1074/jbc.M117.806307 *
STURM ET AL.: "Mitochondrial ATP synthase is dispensable in blood-stage Plasmodium berghei rodent malaria but essential in the mosquito phase", PNAS, vol. 112, no. 33, 2015, pages 10216 - 10223
TRAORE, K. ET AL.: "Do advanced glycation end-products play a role in malaria susceptibility?", PARASITE, vol. 23, no. 15, 2016, pages 1 - 10
ULRIKA WENDEL ET AL: "A novel monoclonal antibody targeting carboxymethyllysine, an advanced glycation end product in atherosclerosis and pancreatic cancer", PLOS ONE, vol. 13, no. 2, 8 February 2018 (2018-02-08), pages e0191872, XP055640398, DOI: 10.1371/journal.pone.0191872 *
VERGNE I ET AL.: "Cell Biology of Mycobacterium tuberculosis Phagosome", ANN REV CELL DEV BIOL., vol. 20, 2004, pages 367 - 94, XP002402783, DOI: 10.1146/annurev.cellbio.20.010403.114015
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, 1987, pages 539,545
VOGEL, F. R. ET AL.: "A compendium of vaccine adjuvants and excipients", PHARMACEUTICAL BIOTECHNOLOGY, vol. 6, 1995, pages 141 - 228
YU, L ET AL.: "Oncogenic virus-induced aerobic glycolysis and tumorigenesis", JOURNAL OF CANCER, vol. 9, no. 20, 2018, pages 3699 - 3706
ZHANG S ET AL.: "Delineation of Diverse Macrophage Activation Programs in Response to Intracellular Parasites and Cytokines", PLOS NEGL TROP DIS, vol. 4, no. 3, 2010, pages e648
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 (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11261241B2 (en) 2008-05-23 2022-03-01 Siwa Corporation Methods, compositions and apparatuses for facilitating regeneration
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)
US11213585B2 (en) 2016-06-23 2022-01-04 Siwa Corporation Vaccines for use in treating various diseases and disorders
US11542324B2 (en) 2017-04-13 2023-01-03 Siwa Corporation Humanized monoclonal advanced glycation end-product antibody
US11958900B2 (en) 2017-04-14 2024-04-16 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
US11518801B1 (en) 2017-12-22 2022-12-06 Siwa Corporation Methods and compositions for treating diabetes and diabetic complications

Also Published As

Publication number Publication date
CN115996954A (en) 2023-04-21
EP4143238A1 (en) 2023-03-08
US20230181730A1 (en) 2023-06-15
JP2023524098A (en) 2023-06-08
IL297799A (en) 2022-12-01
AU2021264007A1 (en) 2022-12-08
CA3177449A1 (en) 2021-11-04

Similar Documents

Publication Publication Date Title
US20230181730A1 (en) Methods of treating infections
US20220175916A1 (en) Methods and compositions for treating chronic effects of radiation and chemical exposure
US10858449B1 (en) Methods and compositions for treating osteoarthritis
US10913797B2 (en) Anti-PD-1 antibodies and therapeutic uses thereof
US9861696B2 (en) Monoclonal antibodies for Ebola and Marburg viruses
US20210253737A1 (en) Methods and compositions for treating disease-related cachexia
TWI697334B (en) Methods for treating allergy and enhancing allergen-specific immunotherapy by administering an il-4r inhibitor
WO2021247397A2 (en) Methods and compositions for enhancing the immune system
JP2021534144A (en) Anti-carboxymethyl lysine antibody and ultrasound to remove AGE-modified cells
US10961321B1 (en) Methods and compositions for treating pain associated with inflammation
JP2021107436A (en) ANTI-TFPI ANTIBODY VARIANTS WITH DIFFERENTIAL BINDING ACROSS pH RANGE FOR IMPROVED PHARMACOKINETICS
AU2017218415A1 (en) Novel anti-LAM and anti-PIM6/LAM monoclonal antibodies for diagnosis and treatment of mycobacterium tuberculosis infections
AU2020264492A1 (en) Recombinant polyclonal proteins and methods of use thereof
WO2022093195A1 (en) Methods and compositions for treating osteoarthritis using anti-age antibodies or age antigens
EA031436B1 (en) Aqueous pharmaceutical composition, pre-filled syringe comprising same and use of the composition in treating autoimmune diseases
KR20170047192A (en) Ebola monoclonal antibodies
WO2023023654A1 (en) Methods and compositions for treating fibrotic diseases
US20130309224A1 (en) Combination of cd37 antibodies with rituximab
US20240000930A1 (en) Methods and compositions for treating kidney diseases
US11518801B1 (en) Methods and compositions for treating diabetes and diabetic complications
US20150231242A1 (en) Combination of cd37 antibodies with bendamustine
US20190336597A1 (en) Methods of generating robust passive and active immune responses
CN116688115B (en) PD-L1/TGF-beta double-function fusion protein preparation and application thereof
KR20230104192A (en) Immune compositions comprising antigens and glycoengineered antibodies thereof

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: 21731330

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 297799

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 3177449

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2022566616

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2021731330

Country of ref document: EP

Effective date: 20221201

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021264007

Country of ref document: AU

Date of ref document: 20210430

Kind code of ref document: A