WO2007022537A2 - Use of lactoferrin as a chemokine and a chemotactic modulator - Google Patents

Use of lactoferrin as a chemokine and a chemotactic modulator Download PDF

Info

Publication number
WO2007022537A2
WO2007022537A2 PCT/US2006/032803 US2006032803W WO2007022537A2 WO 2007022537 A2 WO2007022537 A2 WO 2007022537A2 US 2006032803 W US2006032803 W US 2006032803W WO 2007022537 A2 WO2007022537 A2 WO 2007022537A2
Authority
WO
WIPO (PCT)
Prior art keywords
disease
lactoferrin
composition
cells
wound
Prior art date
Application number
PCT/US2006/032803
Other languages
French (fr)
Other versions
WO2007022537A3 (en
Inventor
Jose Engelmayer
Karel Petrak
Atul Varadhachary
Original Assignee
Agennix Incorporated
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 Agennix Incorporated filed Critical Agennix Incorporated
Publication of WO2007022537A2 publication Critical patent/WO2007022537A2/en
Publication of WO2007022537A3 publication Critical patent/WO2007022537A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/40Transferrins, e.g. lactoferrins, ovotransferrins

Definitions

  • the present invention describes the use of chemotactic modulators to treat diseases with a chemotactic, immune or inflammatory component.
  • the invention also describes the novel use of lactoferrin to modulate the movement or migration of cells and the immune system through its ability to function as a chemotactic modulator by either directly acting as a chemokine or by modulating chemotactic activity either through the induction or inhibition of other chemotactic modulators or by any other means.
  • lactoferrin lactoferrin to modulate the movement or migration of cells and the immune system through its ability to function as a chemotactic modulator by either directly acting as a chemokine or by modulating chemotactic activity either through the induction or inhibition of other chemotactic modulators or by any other means.
  • This constellation of properties to treat diseases with a chemotactic, immune or inflammatory component is envisaged.
  • Lactoferrin is a single chain metal binding glycoprotein. Many cells types, such as epithelial cells, monocytes, macrophages, lymphocytes, and intestinal brush-border cells, are known to have lactoferrin receptors. In addition to lactoferrin being an essential growth factor for both B and T lymphocytes, lactoferrin has a wide array of functions related to host primary defense mechanisms. For example, lactoferrin has been reported to activate natural killer (NK) cells, induce colony-stimulating activity, activate polymorphonuclear neutrophils (PMN), regulate granulopoeisis, enhance antibody-dependent cell cytotoxicity, stimulate lymphokine-activated killer (LAK) cell activity, and potentiate macrophage toxicity.
  • NK natural killer
  • PMN polymorphonuclear neutrophils
  • LAK stimulate lymphokine-activated killer
  • the present invention is directed to a method for modulating the migration and chemokine production of cells by administration of a lactoferrin preparation which may be applied topically, orally or parenterally for treatment of diseases with an immune or inflammatory component.
  • the method involves screening for agents that will inhibit the secretion of chemokines and cytokines or the chemotaxis toward a lactoferrin gradient.
  • a method of treating a disease or condition in a subject comprising the step of administering to the subject a chemotactic modulator composition in an amount sufficient to modulate chemotactic activity in said subject.
  • the disease or condition is selected from the group consisting of a hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
  • chemotactic ⁇ modulator is selected from the group consisting of lactoferrin, chemokine, chemokine receptor, inflammatory or immune mediator, viral anti-chemokine, a mixture of casein and autologous serum, chemotactic C5 fragment, formyl-1-methionyl-l-leucine-l-phenylalanine (f-Met-Leu-Phe), bacterial factor derived from Escherichia coli, the simple peptide formylmethionylalanine, and activated human complement, carbamylcholine, phenylephrine, and cyclic guanosine 5'- monophosphate, nucleosides and nucleotides and prostaglandins
  • a method of treating a disease or condition in a subject comprising the step of administering to a subject a lactoferrin composition in an amount sufficient to modulate chemotactic activity in said subject.
  • chemotactic activity refers to attraction of cells by lactoferrin' s ability to act as a chemokine.
  • chemotactic activity refers to an activity that influences the movement or migration of cells either through the induction or inhibition of other chemotactic modulators.
  • lactoferrin composition comprises an amino acid sequence selected from the group consisting of SEQ.ID.NO.l, SEQ.ID.NO.2, SEQ.ID.NO.3, SEQ.ID.NO.4, SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.il.
  • lactoferrin composition is a topical gel, a solution, or a tablet containing a concentration of about 0.01% to about 20% of lactoferrin.
  • topical gel is composed from the polymers selected from the group of carbopol, carboxymethyl-cellulose, and pluronic polymers.
  • the disease or condition are selected from the group consisting of hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
  • gastrointestinal disorder is Crohn's disease, colitis, necrotizing entercolitis, endometriosis, irritable bowel syndrome, pancreatitis, periodontal disease, and ulcerative colitis.
  • fibrotic disease is selected from the group consisting of diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis, and atherosclerosis.
  • non-infectious inflammatory disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ-specific inflammatory diseases, glomerular disease, Sjogrens disease, Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
  • non-infectious inflammatory disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ-specific inflammatory diseases, glomerular disease, Sjogrens disease, Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
  • autoimmune disease is selected from the group consisting of Active Chronic Hepatitis, Addison's Disease, Anti-phospholipid Syndrome, Atopic Allergy, Atrophic Gastritis, Achlorhydra Autoimmune, Celiac Disease, Crohn's Disease, Cushings Syndrome, Dermatomyositis, (Type I) Diabetes, Discoid Lupus, Erythematosis, Goodpasture's Syndrome, Grave's Disease, Hashimoto's Thyroiditis, Idiopathic Adrenal Atrophy, Idiopathic Thrombocytopenia, Insulin-dependent Diabetes, Lambert-Eaton Syndrome, Lupoid Hepatitis, Lymphopenia, Mixed Connective Tissue Disease, Multiple Sclerosis, Pemphigoid, Pemphigus Vulgaris, Pernicious Anema, Phacogenic Uveitis, Polyarteritis Nodosa, Polyglandular Auto Syndromes, Primary Biliary Cirrhosis, Primary
  • neoplasm is selected from the group consisting of melanoma, non-small cell lung, small-cell lung, lung hepatocarcinoma, retinoblastoma, astrocytoma, gliobastoma, leukemia, neuroblastoma, squamous cell, head, neck, gum, tongue, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, sarcoma, cervical, gastrointestinal, lymphoma, brain, colon, and bladder.
  • hematopoietic neoplasm is selected from the group consisting of acute myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, and chronic lymphocytic leukemia.
  • the hyperproliferative disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, osteoarthritis, leiomyomas, adenomas, lipomas, hemangiomas, fibromas, vascular occlusion, restenosis, atherosclerosis, pre-neoplastic lesions, carcinoma in situ, oral hairy leukoplakia, and psoriasis.
  • the fragile bone disease is selected from the group consisting of osetoporosis, osteitis deformans, osteogenesis imperfecta, osteopetrosis, Rickets, and osteomalacia.
  • a method of treating a disease or condition in a subject by administering to the subject a sufficient amount of a lactoferrin composition, wherein the lactoferrin composition binds to receptors of chemotactic, immune, or inflammatory mediators.
  • chemotactic, immune or inflammatory mediators are chemokines and other chemotactic modulators, cytokines, or growth factors.
  • disease or condition are selected from the group consisting of skin disease, fibrotic disease, chronic inflammatory disease, wound condition, inflammatory condition, autoimmune disease, hyperproliferative disease, fragile bone disease, pain and diabetes mellitus.
  • a method of stimulating a cultured cell to induce chemotactic, inflammatory and/or immune mediators comprising the step of adding a lactoferrin composition to the cell thereby stimulating MIP-lalpha, MlP-l ⁇ , IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP- 3alpha, TECK 5 I-TAC, Lymphotactin, PARC, MIP-I delta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO- ⁇ , GRO- ⁇ , GRO- ⁇ , SCF 5 or MEC/CCL28.
  • FIG. 1 shows lactoferrin acting as a chemokine, a chemokine activity blocker, and a chemokine activity enhancer.
  • FIG. 2 shows attraction of different cell types to lactoferrin, a process that is in part mediated by the IL- 8 receptor.
  • FIG. 3 shows specific binding of lactoferrin to different cell types as assessed by flow cytometry.
  • FIG. 4 shows specific binding of lactoferrin to different cell types as assessed by fluorescent microscopy.
  • FIG. 5 shows specific binding of lactoferrin to de ⁇ nal cells in vivo.
  • FIG. 6 shows a dose response of lactoferrin binding to dermal cells in vivo.
  • FIG. 7 shows specific binding of lactoferrin to receptors for inflammatory or immune mediators.
  • FIG. 8 shows that topical lactoferrin activates the biological activity of inflammatory or immune receptors in vivo in the setting of an open wound.
  • FIG. 9 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human fibroblasts.
  • FIG. 10 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human keratinocytes.
  • FIG. 11 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human hepatocytes.
  • FIG. 12 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in hepatocytes in a time and dose dependent fashion.
  • FIG. 13 shows the release of different inflammatory or immune mediators form hepatocytes upon lactoferrin stimulation.
  • FIG. 14 shows that oral lactoferrin increases the percentage of different immune cells in an immune organ (Peyer's patches) of mice, suggesting that lactoferrin-induced chemotaxis played a role in the process.
  • FIG. 15 shows that oral lactoferrin increases the total number of cells in a mouse immune organ (Peyer's patches), a process that is significantly impaired in IL-8 receptor knockout mice, suggesting that lactoferrin-induced chemoattraction played a role and that this chemotaxis is mediated at least in part by the IL-8 receptor in vivo.
  • FIG. 16 shows that lactoferrin binds to all tested immune cells of a mouse immune organ (spleen), inducing concomitant maturation and/or activation in some cell types.
  • FIG. 17 shows the ability of lactoferrin to bind fibroblasts in vitro by two methods.
  • FIG. 18 shows that lactoferrin attracts normal human dermal fibroblasts from adult, thus functioning as a cheniokine for these cells.
  • FIG.19 shows lactoferrin binding to cells from mouse Peyer's patches with a strong signal peaking early between 1 and 15 minutes after oral administration.
  • FIG. 20 shows that oral lactoferrin induces the migration of labeled dendritic cells into the Peyer's patches
  • acute pain includes tissue injury or pathology.
  • acute pain includes, but is not limited to pain following surgery (e.g., surgery), pain following trauma (e.g., blunt or sharp injury, bullet wounds), pain associated with athletic injury, and the occasional headache.
  • acute pain can have a diagnostic value; that is, it leads to the discovery of a pathological condition. Yet further, acute pain lasts or is anticipated to last for a short period of time, typically less than one to two months.
  • Acute pain is associated with hyperactivity of the sympathetic nervous system, for example, tachycardia, increased respiratory rate, increased blood pressure, increased Cortisol release, diaphoresis and dilated pupils.
  • antibiotic as used herein is defined as a substance that inhibits the growth of microorganisms without damage to the host.
  • the antibiotic may inhibit cell wall synthesis, protein synthesis, nucleic acid synthesis, or alter cell membrane function.
  • Classes of antibiotics that can possibly be used include, but are not limited to, macrolides (e.g., erythromycin), penicillins (e.g., nafcillin), cephalosporins (e.g., cefazolin), carbepenems (e.g., imipenem, aztreonam), other beta-lactam antibiotics, beta-lactam inhibitors (e.g., sulbactam), oxalines (i.e.
  • linezolid aminoglycosides (e.g., gentamicin), chloramphenicol, sufonamides (e.g., sulfamethoxazole), glycopeptides (e.g., vancomycin), quinolones (e.g., ciprofloxacin), tetracyclines (e.g., minocycline), fusidic acid, trimethoprim, metronidazole, clindamycin, mupirocin, rifamycins (e.g., rifampin), streptogramins (e.g., quinupristin and dalfopristin) lipoprotein (e.g., daptomycin), polyenes (e.g., amphotericin B), azoles (e.g., fluconazole), and echinocandins (e.g., caspofungin acetate).
  • anti-depressant drug includes compounds that provide pain relief by reducing side-effects including anxiety.
  • Anti-depressant drugs include, for example, but not limited to tricyclin antidepressants, sedatives, tranquilizers, hypnotics, antihistamines, and amphetamines.
  • the term "antimicrobial” as used herein is defined as a substance that inhibits the growth of microorganisms without damage to the host, for example antibiotics, antifungal and antiseptics.
  • the term "atherosclerosis” as used herein includes a form of arteriosclerosis characterized by a combination of changes in the intima of arteries, such changes include, but are not limited to accumulation of lipids, complex carbohydrates, blood and blood products, fibrous tissue and calcium deposits. Yet further, atherosclerotic plaques can be characterized into at least two areas. One type is characterized by prominent proliferation of cells with small accumulations of lipids. The second type consists mostly of intracellular and extracellular lipid accumulation and a small amount of cellular proliferation.
  • bacteremia as used herein is defined as having a focus of bacterial infection or bacteria in the blood of the subject.
  • cancer as used herein is defined as a hyperproliferation of cells whose unique trait — loss of normal controls — results in unregulated growth, lack of differentiation, local tissue invasion, and metastasis.
  • examples include but are not limited to, melanoma, non-small cell lung, small-cell lung, lung, hepatocarcinoma, leukemia, retinoblastoma, astrocytoma, glioblastoma, gum, tongue, neuroblastoma, head, neck, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, cervical, gastrointestinal, lymphoma, brain, colon, sarcoma or bladder.
  • cardiovascular disease or disorder refers to disease and disorders related to the cardiovascular or circulatory system.
  • Cardiovascular disease and/or disorders include, but are not limited to, diseases and/or disorders of the pericardium, heart valves (e.g., incompetent valves, stenosed valves, rheumatic heart disease, mitral valve prolapse, aortic regurgitation), myocardium (e.g., coronary artery disease, myocardial infarction, heart failure, ischemic heart disease, angina) blood vessels (e.g., hypertension, arteriosclerosis, aneurysm) or veins (e.g., varicose veins, hemorrhoids).
  • heart valves e.g., incompetent valves, stenosed valves, rheumatic heart disease, mitral valve prolapse, aortic regurgitation
  • myocardium e.g., coronary artery disease, myocardial infarction, heart failure, ischemic heart disease,
  • cell The term "cell,” “cell line,” and “cell culture” as used herein may be used interchangeably. All of these terms also include their progeny, which are any and all subsequent generations. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations.
  • cytokine refers to proteins that are made by cells that affect the behavior of other cells, for example stimulate or inhibit cell proliferation.
  • cytokines that are made by lymphocytes are often called lymphokines or interleukins.
  • lymphokines or interleukins are often called lymphokines or interleukins.
  • chemokine refers to small cytokines that are involved in the migration and activation of cells, for example phagocytic cells and lymphocytes.
  • chemokines play a central role in inflammatory and immune response processes.
  • cholesterol refers to the monohydric alcohol form, which is a white, powdery substance that is found in all animal cells and in animal-based foods (not in plants). Cholesterol is an essential nutrient necessary for many functions, including the following: repairing cell membranes, manufacturing vitamin D on the skin's surface, production of hormones, such as estrogen and testosterone, and possibly helping cell connections in the brain that are important for learning and memory.
  • chronic pain refers to pain that lasts longer than 1 month or beyond the resolution of an acute tissue injury or is recurring or is associated with tissue injury and/or chronic diseases that are expected to continue or progress, for example, cancer, arthritis, inflammatory disease, chronic wounds, cardiovascular accidents, spinal cord disorders, central nervous system disorder or recovery from surgery.
  • Chronic pain can be associated with several factors that include, but are not limited to lassitude, sleep disturbance, decreased appetite, loss of taste for food, weight loss, diminished libido, constipation, or depression.
  • chylomicron refers to the largest in size and lowest in density of the triglyceride carrying lipoproteins.
  • diabetes mellitus refers to a disorder of carbohydrate metabolism that is typically characterized by hyperglycemia and glycosuria, which results from inadequate production or utilization of insulin.
  • Diabetes mellitus includes several syndromes or disorders, for example, but not limited to, primary diabetes mellitus (e.g., insulin- dependent (Type I) and non-insulin-dependent (Type II)); secondary diabetes [for example: pancreatic diabetes (e.g., destruction of the pancreas, removal of the pancreas, etc.); extrapancreatic/endocrine diabetes (e.g., hypersomatotropism, hyperadrenalism, hyperthyroidism, glucagonama, etc.); drug-induced diabetes (e.g., steroid diabetes, thiazides, etc.)] and rare/exceptional forms of diabetes (e.g., lipoatrophic diabetes, myatonic diabetes, disturbance of insulin receptors, genetic syndromes, etc).
  • Long-term complications of diabetes include neuropathy, retinopathy, nephropathy, generalized degenerative changes in the blood vessels and increased susceptibility to infection.
  • an effective amount or “therapeutically effective amount” as used herein refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
  • lactoferrin or "LF” as used herein refers to native or recombinant lactoferrin.
  • Native lactoferrin can be obtained T>y purification from mammalian milk or colostrum or from other natural sources.
  • Recombinant lactoferrin (rLF) can be made by recombinant expression or direct production in genetically altered animals, plants, fungi, bacteria, or other prokaryotic or eukaryotic species, or through chemical synthesis.
  • lactoferrin composition refers to a composition having lactoferrin or a part thereof, wherein at least the N-terminal glycine residue is truncated or substituted.
  • N-terminal lactoferrin variant refers to lactoferrin wherein at least the N-terminal glycine has been truncated and/or substituted. N-terminal lactoferrin variants also include, but are not limited to deletion and/or substitution of one or more N-terminal amino acid residues, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 N-terminal amino acid residues, etc. Thus, N-terminal lactoferrin variants comprise at least deletions or truncations and/or substitutions of 1 to 16 N-terminal amino acid residues.
  • the deletion and/or substitution of at least the N-terminal glycine of lactoferrin mediates the same biological effects as full-length lactoferrin and/or may enhance lactoferrin' s biological activity, for example by stimulating the production of various cytokines (i.e., IL-18, MIP-3 ⁇ , GM-CSF or IFN- ⁇ by inhibiting various cytokines, (i.e., IL-2, IL-4, IL-5, IL-IO, and TNF- ⁇ and/or by stimulating.
  • various cytokines i.e., IL-18, MIP-3 ⁇ , GM-CSF or IFN- ⁇
  • various cytokines i.e., IL-2, IL-4, IL-5, IL-IO, and TNF- ⁇ and/or by stimulating.
  • fragment bone condition or “fragile bone disease” as used herein refers to a condition or disease characterized by low bone mass or structural deterioration of bone tissue, leading to bone fragility and increased susceptibility to fractures.
  • gastrointestinal disorder or conditions includes gastrointestinal disorders in which one or more of the symptoms and conditions affect the gastrointestinal tract from the mouth to the anus, as well as any organ that may play a role in digestion, for example, but not limited to pancreas, or gall bladder.
  • Gastrointestinal disorders include, but are not limited to, heartburn, bloating, postoperative ileus, abdominal pain and discomfort, early satiety, epigastric pain, nausea, vomiting, burbulence, regurgitation, intestinal pseudoobstruction, anal incontinence, gastroesophageal reflux disease, irritable bowel syndrome, ulcerative colitis, Crohn's disease, renal disorders, menstrual cramps, pancreatitis, spastic and interstitial cystitis and ulcers and the visceral pain associated therewith.
  • Gram 3 neutropenia refers to the reduction of the absolute neutrophil cell count (ANC) to less than about 1000 cells/ ⁇ L.
  • neutrophil generally refers to a condition in which the ANC is reduced to 1000 cells// ⁇ L or less. Such a condition may be caused by depressed production, increased peripheral destruction of neutrophils.
  • the most common neutropenias are iatrogenic, resulting from the widespread use of cytotoxic or immunosuppressive therapies for cancer treatment or control of autoimmune disorders.
  • neutropenia causes of neutropenia include induction by drugs, hematological diseases including idiopathic, cyclic neutropenia, Chediak-Higashi syndrome, aplastic anemia, infantile genetic disorders, tumor invasion such as myelofibrosis, nutritional deficiency; infections such as tuberculosis, typhoid fever, brucelloisis, tularemia, measles, infectious mononucleosis, malaria, viral hepatitis, leishmaniasis, AIDS, antineutrophil antibodies and/or splenetic or lung trapping, autoimmune disorders, wegner's granulomatosis, acute endotoxemia, hemodialysis, and cardiopulmonary bypass.
  • hematological diseases including idiopathic, cyclic neutropenia, Chediak-Higashi syndrome, aplastic anemia, infantile genetic disorders, tumor invasion such as myelofibrosis, nutritional deficiency
  • infections such as tuberculo
  • graft-versus-host-disease or "GVHD” as used herein is the pathological reaction that occurs between the host and grafted tissue.
  • the grafted or donor tissue dominates the pathological reaction.
  • Graft-versus-host-disease can be seen following stem cell and/or solid organ transplantation. GVHD occurs in immunocompromised subjects, who when transplanted, receive "passenger" lymphocytes in the transplanted stem cells or solid organ. These lymphocytes recognize the recipient's tissue as foreign. Thus, they attack and mount an inflammatory and destructive response in the recipient.
  • GVHD has a predilection for epithelial tissues, especially skin, liver, and mucosa of the gastrointestinal tract.
  • GVHD subjects are immunocompromised due the fact that prior to transplant of the graft, the subject receives immunosuppressive therapy.
  • Gram-negative bacteria or "gram-negative bacterium” as used herein is defined as bacteria which have been classified by the Gram stain as having a red stain. Gram-negative bacteria have thin walled cell membranes consisting of a single layer of peptidoglycan and an outer layer of lipopolysaccharide, lipoprotein, and phospholipid.
  • Exemplary organisms include, but are not limited to, Enterobacteriacea consisting of Escherichia, Shigella, Edwardsiella, Salmonella, Citrobacter, Klebsiella, Enterobacter, Hafnia, Serratia, Proteus, Morganella, Providencia, Yersinia, Erwinia, Buttlauxella, Cedecea, Ewingella, Kluyvera, Tatumella and Rahnella.
  • exemplary gram-negative organisms not in the family Enterobacteriacea include, but are not limited to, Pseudomonas aeruginosa, Stenotrophomonas maltophilia, Burkholderia, Cepacia, Gardenerella, Vaginalis, and Acinetobacter species.
  • Gram-positive bacteria or "gram-positive bacterium” as used herein refers to bacteria, which have been classified using the Gram stain as having a blue stain. Gram-positive bacteria have a thick cell membrane consisting of multiple layers of peptidoglycan and an outside layer of teichoic acid. Exemplary organisms include, but are not limited to, Staphylococcus aureus, coagulase-negative staphylococci, streptococci, enterococci, corynebacteria, and Bacillus species.
  • hyperproliferative disease is defined as a disease that results from a hyperproliferation of cells.
  • hyperproliferative diseases include, but are not limited to cancer or autoimmune diseases.
  • Other hyperproliferative diseases may include vascular occulsion, restenosis, atherosclerosis, or inflammatory bowel disease.
  • immunocompromised as used herein is defined as a subject who is, at the time of pathogen exposure, has a pre-existing condition that reduces one or more mechanisms for normal defense against infection.
  • the immunocompromised condition may be due to a defect or dysfunction of the immune system or to other factors that heighten susceptibility to infection, for example immunosuppressive agents.
  • immunosuppressive agents Although such a categorization allows a conceptual basis for evaluation, immunocompromised individuals with infection often do not fit completely into one group or the other. More than one defect in the body's defense mechanisms may be affected.
  • an immunocompromised state can result from indwelling central lines or other types of impairment due to intravenous drug abuse; or be caused by secondary malignancy, malnutrition, or having been infected with other infectious agents such as tuberculosis, influenza, Staphylococcus aureus or sexually transmitted diseases, e.g., syphilis or hepatitis.
  • insulin-dependent diabetes mellitus refers to diabetes that is characterized by a hyperglycemia, glycosuria, and low blood insulin levels. Type I diabetes can develop at any age. It typically has an abrupt onset during the first two decades of life. Insulin therapy is usually required.
  • intermediate density lipoprotein refers to a triglyceride-carrying lipoprotein.
  • lipid refers to the building blocks of any of the fats or fatty substances found in animals and plants, which are characterized by their insolubility in water and solubility in fat solvents such as alcohol, ether and chloroform. Lipids include fats (e.g., esters of fatty acids and glycerol); lipoids (e.g., phospholipids, cerebrosides, waxes) and sterols (e.g., cholesterol).
  • lipoproteins as used herein are protein spheres that transport cholesterol, triglyceride, or other lipid molecules through the bloodstream. Lipoproteins are categorized into five types according to size and density. They can be further defined by whether they carry cholesterol [the two smaller lipoproteins (HDL and LDL)] or triglycerides [the three largest lipoproteins (IDL, VLDL, and chylomicrons)].
  • LDL low density lipoprotein
  • metal chelator refers to a compound which binds metal.
  • Metal chelators that can be used in the present invention include the divalent metal chelators, for example, ethylenediaminetetraacetic acid (EDTA), [ethylenebis (oxyethylenenitrilo)] tetraacetic acid (EGTA), l,2-bis(2-aminophenoxy)ethane-N,N,N',N'- tetraacetic acid (BAPTA), hydroxyethlene triamine diacetic acid, (HEDTA) or salts thereof.
  • EDTA ethylenediaminetetraacetic acid
  • EGTA ethylenebis (oxyethylenenitrilo)] tetraacetic acid
  • BAPTA l,2-bis(2-aminophenoxy)ethane-N,N,N',N'- tetraacetic acid
  • HEDTA hydroxyethlene triamine diacetic acid
  • morbidity is the state of being diseased. Yet further, morbidity can also refer to the disease rate or the ratio of sick subjects or cases of disease in to a given population.
  • the term "mortality” as used herein is the state of being mortal or causing death. Yet further, mortality can also refer to the death rate or the ratio of number of deaths to a given population.
  • neurode refers to conditions, disorders, and/or diseases that are associated with the nervous system.
  • any condition, disorder and/or disease that effects any component or aspect of the nervous system is referred to as a neurological condition, disorder and/or disease.
  • the term “neurological” or “neurology” encompasses the terms “neuropsychiatric” or “neuropsychiatry” and “neuropsychological” or “neuropsychological”.
  • a neurological disease, condition, or disorder includes, but is not limited to cognitive disorders, affective disorders (e.g., depression disorders and/or anxiety disorders), movement disorders, mental disorders, pain disorders, sleep disorders, etc.
  • neutropenia refers to an a decrease or small number of neutrophils in the blood compared to normal.
  • the World Health Organization defines neutropenia as a subject having an absolute neutrophil cell count (ANC) of about 2000 cells/ ⁇ L or less.
  • ANC absolute neutrophil cell count
  • a subject suffering from neutropenia is one having an ANC of about 2000 cells/ ⁇ L or less, for example 1000 cells/ ⁇ L or even less than 500 cells/ ⁇ L.
  • non-insulin-dependent diabetes mellitus refers to diabetes that is characterized by hyperglycemia and insulin levels being normal to high.
  • Type II diabetes is a form of diabetes mellitus that has gradual onset in obese individuals over the age of 35. Insulin therapy is usually not required; however, Type II diabetes can be destined or prone to become fully insulin-dependent.
  • organ or tissue transplant rejection refers to a consequence of organ or tissue transplantation caused by the recipient's or host's immune system in response to the transplanted organ/tissue, which can damage or destroy it.
  • organ or tissue transplant rejection is controlled by the host subject.
  • osteoporosis as used herein is defined as a general term for describing any disease process that results in reduction in the mass of bone.
  • pain refers to an unpleasant sensation.
  • the subject experiences discomfort, distress or suffering. Pain of a moderate or high intensity is typically accompanied by anxiety.
  • Pain may have dual properties, for example sensation and emotion.
  • parenteral administration includes any form of administration in which the compound is absorbed into the subject without involving absorption via the intestines.
  • exemplary parenteral administrations that are used in the present invention include, but are not limited to intramuscular, intravenous, intraperitoneal, intraoccular, or intraarticular administration. Yet further, parenteral administration also includes administration into a surgical field.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the vectors or cells of the present invention, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
  • the term "pharmaceutical composition” as used herein refers to a lactoferrin composition that this dispersed in a pharmaceutically acceptable carrier.
  • the lactoferrin composition can comprise lactoferrin or an N-terminal lactoferrin variant in which at least the N-terminal glycine amino acid residue is truncated or substituted.
  • oral administration includes, but is not limited to oral, buccal, enteral or intragastric administration.
  • subject as used herein, is taken to mean any mammalian subject to which a human lactoferrin composition is orally administered according to the methods described herein.
  • the methods of the present invention are employed to treat a human subject.
  • Another embodiment includes treating a human subject suffering from a wound.
  • terapéuticaally effective amount refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
  • topical administration includes, but is not limited to topical, dermal (e.g., trans-dermal or intra-dermal), epidermal, or subcutaneous.
  • treating and “treatment” as used herein refers to administering to a subject a therapeutically effective amount of a recombinant human lactoferrin composition so that the subject has an improvement in the disease.
  • the improvement is any improvement or remediation of the symptoms.
  • the improvement is an observable or measurable improvement.
  • a treatment may improve the disease condition, but may not be a complete cure for the disease.
  • wound refers to any injury, such as an ulcer, as a result of disease or disorder, or as a result of an accident, incident, or surgical procedure. Wound can be further defined as acute and/or chronic.
  • preventing refers to minimizing, reducing or suppressing the risk of developing a disease state or parameters relating to the disease state or progression or other abnormal or deleterious conditions.
  • respiratory disorder refers to any condition and/or disorder relating to respiration and/or the respiratory system.
  • the respiratory disorder can be an allergic or non-allergic respiratory disorder. More specifically, the respiratory disorder includes, but is not limited to atopic asthma, non-atopic asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis and allergic rhinitis.
  • spontaneous infection is defined as a Systemic Inflammatory Response Syndrome to an infective process in which severe derangement of the host immune system fails to prevent extensive 'spill over' of inflammatory mediators from a local infection focus into the systemic circulation.
  • septic shock as used herein is a consequence of sepsis in which the systemic inflammatory response leads to the failure of vital organs' function (for example of the lungs as in ARDS).
  • the term "subject” as used herein, is taken to mean any mammalian subject to which a lactoferrin composition is administered according to the methods described herein.
  • a mammalian subject includes, but is not limited to humans, monkeys, horses, pigs, cows, dogs, cats, rats and mice.
  • the methods of the present invention are employed to treat a human subject.
  • the term "Th2 cells” as used herein is defined as a subset of CD4 T-cells that are characterized by the cytokines they produce. These cells are mainly involved in stimulating B cells to produce antibody and are often called helper T-cells. It is also known that extracellular antigens tend to stimulate the production of Th2 cells. Thus, as used herein, "Th2 cells” is interchangeable with "helper T-cells”.
  • total cholesterol refers to the sum of three kinds of lipids: high-density lipoprotein (HDL), low-density lipoprotein (LDL), and triglycerides.
  • HDL high-density lipoprotein
  • LDL low-density lipoprotein
  • triglycerides levels of serum total cholesterol of > 200 mg/dl are levels that are an indicating risk factor for atherosclerosis and cardiovascular disease.
  • triglycerides as used herein are composed of fatty acid molecules and are the basic chemicals contained in fats in both animals and plants.
  • type I osteoporosis or "postmenopausal osteoporosis” as used herein is usually found in women after the beginning of menopause. The incidence in women is six to eight times higher than that in men. It has been postulated that the cause of this osteoporosis is accelerated bone resorption. The increased bone turnover results in a secondary decrease in parathyroid hormone (PTH) secretion as well as a secondary reduction in the renal production of calcitriol. Patients present with trabecular bone loss with vertebral fractures or distal forearm fractures.
  • PTH parathyroid hormone
  • osteoporosis or "age-associated osteoporosis” or “senile osteoporosis” occurs in men or women over the age of 70.
  • the mechanisms of this bone mass loss are thought to be increased PTH secretion resulting from decreased gastrointestinal calcium absorption and decreased osteoblast function.
  • Patients usually present with fractures of the hip or vertebrae, sites that contain cortical and trabecular bone, although fractures of the pelvis, ribs, and tibia can also occur.
  • VLDL very low density lipoprotein
  • the lactoferrin used according to the present invention can be obtained through isolation and purification from natural sources, for example, but not limited to mammalian milk.
  • the lactoferrin is preferably mammalian lactoferrin, such as bovine or human lactoferrin.
  • the lactoferrin is produced recombinantly using genetic engineering techniques well known and used in the art, such as recombinant expression or direct production in genetically altered animals, plants or eukaryotes, or chemical synthesis. See, i.e., U.S. Patent Nos. 5,571,896; 5,571,697 and 5,571,691, which are herein incorporated by reference.
  • the present invention provides lactoferrin variants having enhanced biological activities of natural LF and or rLF, e.g., the ability to stimulate and/or inhibit cytokines or chemokines or the ability of lactoerrin and/or its variants to function as a chemokine.
  • the invention provides variants of lactoferrin from which at least the N-terminal glycine residue has been substituted and/or truncated.
  • the N-terminal lactoferrin variants may occur naturally or may be modified by the substitution or deletion of one or more amino acids.
  • the present invention concerns novel peptide compositions.
  • a "peptide,” “amino acid molecule,” “polypeptide”, “peptide composition” generally refers, but is not limited to, a peptide of about 33 amino acids.
  • all the terms related "peptide” are interchangeable.
  • a lactoferrin related peptide includes related-compounds of the respective molecules that exhibit at least some biological activity in common with their native counterparts, for example lactoferrin.
  • Such related-compounds include, but are not limited to, truncated polypeptides and polypeptides having fewer amino acids than the native polypeptide.
  • the full length lactoferrin protein which includes polypeptide sequences, for example, but are not limited to SEQ.ID.NO.12 (GenBank accession AAA36159.1), SEQ.ID.NO.13 (GenBank accession CAA38572.1); SEQ.ID.NO.14 (GenBank accession NP_032548.2); SEQ.ID.NO.15 (GenBank accession NP_002334.1); SEQ.ID.NO.16 (GenBank accession NP_851341.1); SEQ.ID.NO.17 (GenBank accession AANl 1304.1); SEQ.ID.NO.18 (GenBank accession AAN75578.2); SEQ.ID.NO.19 (GenBank accession CAA06441.1); SEQ.ID.NO.20 (GenBank accession BAA13633.1); SEQ.ID.NO.21 (GenBank accessionAAG48753.1); SEQ.ID.NO.22 (GenBank accession BAB03470.1); SEQ.ID.NO.23 (GenBank accession
  • lactoferrin related peptides may include the following sequences, but are not limited to these sequences, SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP-31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO;8 (LFP-33), SEQJD.NO.9 (LFP-34), SEQ.ID.NO.IO (LFP-35), and SEQ.ID.NO.i l (LFP-36).
  • SEQ.ID.NO.l LFP-21
  • SEQ.ID.NO.2 LFP-22
  • SEQ.ID.NO.3 LFP-23
  • SEQ.ID.NO.4 LFP-24
  • SEQ.ID.NO.5 LFP-25
  • the size of the peptide composition or amino acid molecule may comprise, but is not limited to, about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 60 or greater amino molecule residues, and any range derivable therein.
  • an "amino molecule” refers to any amino acid, amino acid derivative or amino acid mimic as would be known to one of ordinary skill in the art.
  • the residues of the peptide are sequential or contiguous, without any non-amino molecule interrupting the sequence of amino molecule residues.
  • the sequence may comprise one or more non-amino molecule moieties.
  • the sequence of residues of the peptide may be interrupted by one or more non-amino molecule moieties.
  • peptide encompasses amino molecule sequences comprising at least one of the 20 common amino acids in naturally synthesized proteins, or at least one modified or unusual amino acid, including but not limited to those shown on Table 1 below.
  • Peptide compositions may be made by any technique known to those of skill in the art, including the expression of proteins, polypeptides or peptides through standard molecular biological techniques, the isolation of peptide compounds from natural sources, or the chemical synthesis of peptides.
  • the nucleotide and protein, polypeptide and peptide sequences for various genes have been previously disclosed, and may be found at computerized databases known to those of ordinary skill in the art.
  • One such database is the National Center for Biotechnology Information's Genbank and GenPept databases.
  • the coding regions for these known genes may be amplified and/or expressed using the techniques disclosed herein or as would be know to those of ordinary skill in the art.
  • various commercial preparations of polypeptides and peptides are known to those of skill in the art.
  • a peptide composition may be purified.
  • purified will refer to a specific polypeptide, or peptide composition that has been subjected to fractionation to remove various other proteins, polypeptides, or peptides, and which composition substantially retains its activity, as may be assessed, for example, by the protein assays, as would be known to one of ordinary skill in the art for the specific or desired polypeptide or peptide.
  • Deletional variants can be produced by proteolysis of lactoferrin and/or expression of a polynucleotide encoding a truncated lactoferrin as described in U.S. Patent 6,333,311, which is incorporated herein by reference.
  • Substitutional variants or replacement variants typically contain the exchange of one amino acid for another at one or more sites within the protein. Substitutions can be conservative, that is, one amino acid is replaced with one of similar shape and charge. Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyros
  • Substitutional variants or replacement can be produced using Standard mutagenesis techniques, for example, site-directed mutagenesis as disclosed in U.S. Patents 5,220,007; 5,284,760; 5,354,670; 5,366,878; 5,389,514; 5,635,377; 5,789,166, and 6,333,311, which are incorporated herein by reference.
  • the hydropathic index of amino acids may be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics (Kyte and Doolittle, 1982), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (- 1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, e.g., still obtain a biological functionally equivalent protein.
  • substitution of amino acids whose hydropathic indices are within ⁇ 2 is preferred, those that are within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • Patent 4,554,101 the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ⁇ 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4).
  • an amino acid can be substituted for another having a similar hydrophilicity value and still obtains a biologically equivalent and immunologically equivalent protein.
  • substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those that are within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • variants can include variants in which at least the N-terminal glycine amino acid residue can be replaced or substituted with any of the twenty natural occurring amino acids, for example a positively charged amino acid (arginine, lysine, or histidine), a neutral amino acid (alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylaline, proline, serine, threonine, tryptophan, tyrosine, valine) and/or a negatively charged amino acid (aspartic acid or glutamic acid).
  • a positively charged amino acid arginine, lysine, or histidine
  • a neutral amino acid alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylaline, proline, serine, threonine, tryptophan, tyrosine, valine
  • any amino acid residue within the range of Nl to Nl 6 can be replaced or substituted. It is envisioned that at least up to 16 of the N-terminal amino acids residues can be replaced or substituted as long as the protein retains it biological and/or functional activity, which is stimulating the production of various cytokines, such as IL-18, MIP-3 ⁇ , GM-CSF or IFN- ⁇ , or inhibiting production of various cytokines, such as IL-2, IL-4, IL-5, IL-10, and TNF- ⁇ .
  • the N-terminal lactoferrin variants of the present invention are considered functional equivalents oflactoferrin.
  • N-terminal amino acid residues can be substituted with a modified and/or unusual amino acids.
  • a table of exemplary, but not limiting, modified and/or unusual amino acids is provided herein below.
  • N-terminal lactoferrin variants differs and/or substitutions
  • lactoferrin composition may be done by determination of the N-terminal amino acid sequence by the process of Edman degradation using standard methods.
  • a relative proportion of N-terminal lactoferrin variant comprises at least 1% of the lactoferrin composition, at least 5% of the lactoferrin composition, at least 10% of the lactoferrin composition, at least 25% of the lactoferrin composition, at least 50% of the lactoferrin composition or any range in between.
  • the protein is reacted with phenylisothiocyanate (PITC), which reacts with the amino acid residue at the amino terminus under basic conditions to form a phenylthiocarbamyl derivative (PTC-protein).
  • PITC phenylisothiocyanate
  • Trifluoroacetic acid then cleaves off the first amino acid as its anilinothialinone derivative (ATZ-amino acid) and leaves the new amino terminus for the next degradation cycle.
  • N-terminal lactoferrin variant may also be done more precisely by using a Dansylation reaction. Briefly, protein is dansylated using dansyl chloride reacted with the protein in alkaline conditions (pH 10). Following the Dansylation, the reaction mixtures are dried to pellets, then completely hydrolyzed in 6N HCl. The proportion of N- terminal amino acids are identified by RP HPLC using an in-line fluorometer in comparison with standards made up of known dansylated amino acids.
  • biologically functional equivalents it is well understood by the skilled artisan that, inherent in the definition of a "biologically functional equivalent" protein is the concept that there is a limit to the number of changes that may be made within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity and/or enhancing the biological activity of the lactoferrin molecule.
  • Biologically functional equivalents are thus defined herein as those proteins in which selected amino acids (or codons) may be substituted.
  • Functional activity is defined as the ability of the lactoferrin related peptide to stimulate or inhibit various cytokines or chemokines.
  • lactoferrin peptides of this invention can be in glycosylated or unglycosylated form, can be modified post-translationally (e.g., acetylation, and phosphorylation) or can be modified synthetically (e.g., the attachment of a labeling group). Fragments of lactoferrin molecules that retain the prophylactic efficacy can also be used (see, e.g. PCT/IB 00/00271, which is incorporated herein in reference in its entirety).
  • the present invention also describes lactoferrin, lactoferrin variants and lactoferrin related peptides for use in various embodiments of the present invention. Because of their relatively small size, the peptides of the invention can also be synthesized in solution or on a solid support in accordance with conventional techniques. Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, for example, Stewart and Young (1984); Tarn et al. (1983); Merrifield (1986); and Barany and Merrifield (1979), each incorporated herein by reference.
  • Short peptide sequences or libraries of overlapping peptides, usually from about 6 up to about 35 to 50 amino acids, which correspond to the selected regions described herein, can be readily synthesized and then screened in screening assays designed to identify reactive peptides.
  • recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a peptide of the invention is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression.
  • Certain embodiments of the present invention concern a nucleic acid composition having a nucleic acid sequence of lactoferrin.
  • a lactoferrin nucleic acid comprises a wild-type or a mutant lactoferrin nucleic acid.
  • a lactoferrin nucleic acid sequence encodes for or comprises a transcribed nucleic acid.
  • a lactoferrin nucleic acid sequence comprises a nucleic acid segment of SEQ.ID.NO.40 (GenBank accession M93150); SEQ.ID.NO.41 (GenBank accession X54801); SEQ.ID.NO.42 (GenBank accession NM_008522); SEQ.ID.NO.43 (GenBank accession NM_002343); SEQ.ID.NO.44 (GenBank accession NM_180998); SEQ.ID.NO.45 (GenBank accession A ⁇ 137470); SEQ.ID.NO.46 (GenBank ⁇ accession AYl 78998); " SEQ ⁇ JD.NO.47 ⁇ (GenBank accession AJ005203); SEQ.ID.NO.48 (GenBank accession D88510); SEQ.ID.NO.49 (GenBank accession AF332168); SEQ.ID.NO.50 (GenBank accession AB046664); SEQ.ID.NO.51 (GenBank accession AAF
  • nucleic acid sequence of the present invention encodes a polypeptide, peptide, for example, but not limited to SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP- 31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO.8 (LFP-33), SEQ.ID.NO.9 (LFP-34), SEQ.ID.NO.10 (LFP-35), and SEQ.ID.NO.i l (LFP-36).
  • SEQ.ID.NO.l LFP-21
  • SEQ.ID.NO.2 LFP-22
  • SEQ.ID.NO.3 LFP-23
  • SEQ.ID.NO.4 LFP-24
  • SEQ.ID.NO.5 LFP-25
  • nucleic acid compositions encoding lactoferrin and/or lactoferrin related peptides are herein provided and are also available to a skilled artisan at accessible databases, including the National Center for Biotechnology Information's GenBank database and/or commercially available databases, such as from Celera Genomics, Inc. (Rockville, MD).
  • nucleic acid is well known in the art.
  • a “nucleic acid” as used herein will generally refer to a molecule (i.e., a strand) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase.
  • a nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine "A,” a guanine "G,” a thymine “T” or a cytosine “C”) or RNA (e.g., an A, a G, an uracil "U” or a C).
  • nucleic acid encompass the terms “oligonucleotide” and “polynucleotide,” each as a subgenus of the term “nucleic acid.”
  • oligonucleotide refers to a molecule of between about 3 and about 100 nucleobases in length.
  • polynucleotide refers to at least one molecule of greater than about 100 nucleobases in length.
  • a nucleic acid may encompass a double-stranded molecule or a triple-stranded molecule that comprises one or more complementary strand(s) or "complement(s)" of a particular sequence comprising a molecule.
  • a single stranded nucleic acid may be denoted by the prefix "ss,” a double stranded nucleic acid by the prefix "ds,” and a triple stranded nucleic acid by the prefix "ts.”
  • nucleic acids of the present invention encompass biologically functional equivalent lactoferrin proteins, polypeptides, or lactoferrin related peptides. Such sequences may arise as a consequence of codon redundancy or functional equivalency that is known to occur naturally within nucleic acid sequences or the proteins, polypeptides or peptides thus encoded.
  • functionally equivalent proteins, polypeptides or peptides may be created via the application of recombinant DNA technology, in which changes in the protein, polypeptide or peptide structure may be engineered, based on considerations of the properties of the amino acids being exchanged. Changes designed by man may be introduced, for example, through the application of site-directed mutagenesis techniques as discussed herein below, e.g., to introduce improvements or alterations to the antigenicity of the protein, polypeptide or peptide.
  • the present invention may involve using expression constructs as the pharmaceutical composition.
  • the expression construct comprises polynucleotide sequences encoding polypeptides which can enhance the immune system, for example the expression construct may comprise a nucleic acid sequence encoding the amino acid sequence of SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP- 31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO.8 (LFP-33), SEQ.ID.NO.9 (LFP-34), SEQ.ID.NO.10 (LFP-35), SEQ.ID.NO.i l (LFP-36), SEQ.ID.NO.12.
  • the present invention involves the manipulation of genetic material to produce expression constructs that encode the peptides of the present invention.
  • the peptides are contained in an expression vector.
  • Such methods involve the generation of expression constructs containing, for example, a heterologous nucleic acid sequence encoding the peptide of interest and a means for its expression, replicating the vector in an appropriate cell, obtaining viral particles produced therefrom, and infecting cells with the recombinant virus particles.
  • expression vector refers to any type of genetic construct comprising a nucleic acid coding for a lactoferrin related peptide, for example, but not limited to SEQ.ID.NO.l, SEQ.ID.NO.2, SEQ.ID.NO.3, SEQ.ID.NO.4, SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.i l.
  • DNA molecules are then translated into a protein, polypeptide, or peptide.
  • Expression vectors can contain a variety of "control sequences," which refer to nucleic acid sequences necessary for the transcription and possibly translation of an operably linked coding sequence in a particular host cell.
  • vectors and expression vectors may contain nucleic acid sequences that serve other functions as well and are described infra. It is contemplated in the present invention, that virtually any type of vector may be employed in any known or later discovered method to deliver nucleic acids encoding a lactoferrin and/or lactoferrin related peptide.
  • An expression vector comprising a nucleic acid encoding a lactoferrin and/or lactoferrin related peptide may comprise a virus or engineered construct derived from a viral genome.
  • a plasmid vector is contemplated for use to transform a host cell.
  • plasmid vectors containing replicon and control sequences that are derived from species compatible with the host cell are used in connection with these hosts.
  • the vector ordinarily carries a replication site, as well as marking sequences that are capable of providing phenotypic selection in transformed cells.
  • Plasmid vectors are well known and are commercially available. Such vectors include, but are not limited to, the commercially available pSupervector (OligoEngine, Seattle, WA), and pSuppressorNeo vector (IMGENEX Corporation).
  • pREP series of vectors such as the pREP series of vectors, pBPV, pMSG, pSVL (Pharmacia), adenovirus vector (AAV; pCWRSV, Chatterjee et al.
  • retroviral vectors such as the pBABE vector series, a retroviral vector derived from MoMuLV (pGINa, Zhou et al., (1994)); and pTZ18U (BioRad, Hercules, CA).
  • a gene encoding a lactoferrin and/or lactoferrin related peptide is introduced in vivo in a viral vector.
  • the ability of certain viruses to enter cells via receptor-mediated endocytosis and to integrate into the host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells (Ridgeway, 1988; Nicolas and Rubenstein, 1988; Baichwal and Sugden, 1986; Temin, 1986).
  • Such vectors include an attenuated or defective DNA virus, such as but not limited to herpes simplex virus (HSV), papilloma virus, Epstein Barr virus (EBV), adenovirus, adeno-associated virus (AAV), lentivirus and the like.
  • HSV herpes simplex virus
  • EBV Epstein Barr virus
  • AAV adeno-associated virus
  • lentivirus lentivirus
  • Defective viruses which entirely or almost entirely lack viral genes, are preferred. Defective virus is not infective after introduction into a cell.
  • Use of defective viral vectors allows for administration to cells in a specific, localized area, without concern that the vector can infect other cells. Thus, any tissue can be specifically targeted.
  • vectors examples include, but are not limited to, a defective herpes virus 1 (HSVl) vector (Kaplitt et al., 1991) an attenuated adenovirus vector, (Stratford- Perricaudet et al., 1992), a defective adeno-associated virus vector (Samulski et al., 1987 and Samulski et al., 1989) or pox- vector.
  • HSVl herpes virus 1
  • Such vectors may be used to (i) transform cell lines in vitro for the purpose of expressing the a lactoferrin and/or lactoferrin related peptide or (ii) to transform cells in vitro or in vivo to provide therapeutic molecules for gene therapy.
  • the present invention contemplates viral vectors such as, but not limited to, an adenoviral vector, an adeno-associated viral vector, a retroviral vector, a lentiviral vector, a herpes viral vector, polyoma viral vector, pox- vector or hepatitis B viral vector.
  • viral vectors such as, but not limited to, an adenoviral vector, an adeno-associated viral vector, a retroviral vector, a lentiviral vector, a herpes viral vector, polyoma viral vector, pox- vector or hepatitis B viral vector.
  • an appropriate immunosuppressive treatment is employed in conjunction with the viral vector, e.g., adenovirus vector, to avoid immunodeactivation of the viral vector and transfected cells.
  • the viral vector e.g., adenovirus vector
  • immunosuppressive cytokines such as interleukin-12 (IL- 12), interferon- ⁇ (IFN- ⁇ ), or anti-CD4 antibody
  • IL-12 interleukin-12
  • IFN- ⁇ interferon- ⁇
  • anti-CD4 antibody can be administered to block humoral or cellular immune responses to the viral vectors (Wilson, Nature Medicine (1995).
  • the gene can be introduced in a retroviral vector, e.g., as described in U.S. Pat. No. 5,399,346; Mann et al., 1983; U.S. Pat. No. 4,650,764; U.S. Pat. No. 4,980,289; Markowitz et al., 1988; U.S. Pat. No. 5,124,263; International Patent Publication No7 WO 95707358; and Kuo et al., 1993, each of which is incorporated herein by reference in its entirety.
  • Targeted gene delivery is described in International Patent Publication WO 95/28494.
  • the vector can be introduced in vivo by lipofection.
  • liposomes for encapsulation and transfection of nucleic acids in vitro.
  • Synthetic cationic lipids designed to limit the difficulties and dangers encountered with liposome mediated transfection can be used to prepare liposomes for in vivo transfection of a gene encoding a marker.
  • the use of cationic lipids may promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes.
  • the use of lipofection to introduce exogenous genes into the specific organs in vivo has certain practical advantages. Molecular targeting of liposomes to specific cells represents one area of benefit.
  • Lipids may be chemically coupled to other molecules for the purpose of targeting.
  • Targeted peptides e.g., hormones or neurotransmitters, and proteins such as antibodies, or non-peptide molecules could be coupled to liposomes chemically.
  • cell As used herein, the terms “cell,” “cell line,” and “cell culture” may be used interchangeably. All of these terms also include their progeny, which is any and all subsequent generation formed by cell division. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations.
  • a host cell may be "transfected” or “transformed,” which refers to a process by which exogenous nucleic acid is transferred or introduced into the host cell.
  • a transformed cell includes the primary subject cell and its progeny.
  • the terms “engineered” and “recombinant” cells or host cells are intended to refer to a cell into which an exogenous nucleic acid sequence, such as, for example, a lactoferrin and/or lactoferrin related peptide. Therefore, recombinant cells are distinguishable from naturally occurring cells that do not contain a recombinantly introduced nucleic acid.
  • Host cells may be derived from prokaryotes or eukaryotes, depending upon whether the desired result is replication of the vector or expression of part or all of the vector encoded nucleic acid sequences. Numerous cell lines and cultures are available for use as a host cell, and they can be obtained through the American Type Culture Collection (ATCC), which is an organization that serves as an archive for living cultures and genetic materials.
  • ATCC American Type Culture Collection
  • a cell may comprise, but is not limited to, at least one skin, bone, neuron, axon, cartilage, blood vessel, cornea, muscle, facia, brain, prostate, breast, endometrium, lung, pancreas, small intestine, blood, liver, testes, ovaries, cervix, colon, skin, stomach, esophagus, spleen, lymph node, bone marrow, kidney, peripheral blood, embryonic or ascite cell, and all cancers thereof.
  • An appropriate host can be determined by one of skill in the art based on the vector backbone and the desired result.
  • Bacterial cells used as host cells for vector replication and/or expression include DH5 ⁇ , JMl 09, and KC8, as well as a number of commercially available bacterial hosts such as SURE® Competent Cells and SOLOP ACKTM Gold Cells (STRATAGENE®, La JoUa).
  • Examples of eukaryotic host cells for replication and/or expression of a vector include HeLa, NIH3T3, Jurkat, 293, Cos, CHO, Saos, and PC12.
  • Other eukaryotic cells include yeast cells, such as Aspergillus species and Saccharomyces species. Many host cells from various cell types and organisms are available and would be known to one of skill in the art.
  • a host cell strain may be chosen that modulates the expression of the inserted sequences, and/or modifies and/or processes the gene product in the specific fashion desired.
  • modifications e.g., glycosylation
  • processing e.g., cleavage
  • protein products may be important for the function of the protein.
  • the present invention is drawn to a composition comprising a lactoferrin composition that is dispersed in a pharmaceutical carrier.
  • the lactoferrin that is contained in the composition of the present invention comprises lactoferrin or an N-terminal lactoferrin variant in which at least the N-I terminal glycine residue is truncated or substituted. More specifically, the N-terminal lactoferrin variant comprises at least 1% of the composition, at least 5% of the composition, at least 10% of the composition, at least 25% of the composition, at least 50% of the composition or any range in between.
  • lactoferrin that is contained in the composition of the present invention comprises lactoferrin related peptides, for example, SEQ.ID.NOr 1, SEQ.ID.NO.2, SEQ.ID.NO.3 ⁇ SEQTD.N ⁇ . V SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.i l and/or expression vectors encoding a lactoferrin related peptide.
  • lactoferrin related peptides for example, SEQ.ID.NOr 1, SEQ.ID.NO.2, SEQ.ID.NO.3 ⁇ SEQTD.N ⁇ . V SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.i
  • the composition comprises lactoferrin in combination with a metal chelator dispersed in a pharmaceutical carrier.
  • a metal chelator that is dispersed in a pharmaceutical carrier.
  • both compositions e.g., lactoferrin alone or lactoferrin in combination with a metal chelator
  • the addition of a metal chelator to the lactoferrin composition enhances the sequestering of metal ions and thus strengthens the immune system or enhances the effect of lactoferrin.
  • Metal chelators that can be used in combination with lactoferrin include the divalent metal chelators, for example, ethylenediaminetetraacetic acid (EDTA), [ethylenebis(oxyethylenenitrilo)] tetraacetic acid (EGTA), l,2-bis(2-aminophenoxy)ethane- N,N,N',N'-tetraacetic acid (BAPTA), hydroxyethlene triamine diacetic acid, (HEDTA) or any salts thereof. More preferably, EDTA is used in combination with lactoferrin.
  • EDTA ethylenediaminetetraacetic acid
  • EGTA ethylenebis(oxyethylenenitrilo)] tetraacetic acid
  • BAPTA l,2-bis(2-aminophenoxy)ethane- N,N,N',N'-tetraacetic acid
  • HEDTA hydroxyethlene triamine diacetic acid
  • the present invention may concern the use of a pharmaceutical lipid vehicle compositions that include lactoferrin or lactoferrin related peptides, one or more lipids, and an aqueous solvent.
  • lipid will be defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds are well known to those of skill in the art, and as the term "lipid” is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man). However, a lipid is usually a biological substance.
  • Biological lipids are well known in the art, and include for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • neutral fats phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • lipids are also encompassed by the compositions and methods of the present invention.
  • the lactoferrin or lactoferrin related peptides may be dispersed in a solution containing a lipid, dissolved with a lipid, emulsified with a lipid, mixed with a lipid, combined with a lipid, covalently bonded to a lipid, contained as a suspension in a lipid, contained or complexed with a micelle or liposome, or otherwise associated with a lipid or lipid structure by any means known to those of ordinary skill in the art.
  • the dispersion may or may not result in the formation of liposomes.
  • composition for oral administration which is combined with a semisolid or solid carrier can be further formulated into hard or soft shell gelatin capsules, tablets, or pills. More preferably, gelatin capsules, tablets, or pills are enterically coated. Enteric coatings prevent denaturation of the composition in the stomach or upper bowel where the pH is acidic. See, e.g., U.S. Pat. No. 5,629,001. Upon reaching the small intestines, the basic pH therein dissolves the coating and permits the lactoferrin composition to be released and absorbed by specialized cells, e.g., epithelial enterocytes and Peyer's patch M cells.
  • specialized cells e.g., epithelial enterocytes and Peyer's patch M cells.
  • Oral administration of the composition includes oral, buccal, enteral or intragastric administration. It is also envisioned that the composition may be used as a food additive. For example, the composition is sprinkled on food or added to a liquid prior to ingestion. In further embodiments, the oral composition is administered in conjunction with an antacid. Thus, an antacid is administered prior or substantially simultaneously with or after oral administration of the composition. The administration of an antacid just prior or immediately following the administration of the composition may help to reduce the degree of inactivation of the lactoferrin in the digestive tract. Examples of appropriate antacids include, but are not limited to, sodium bicarbonate, magnesium oxide, magnesium hydroxide, calcium carbonate, magnesium trisilicate, magnesium carbonate and alumin hydroxide gel.
  • a powdered composition is combined with a liquid carrier such as, i.e., water or a saline solution, with or without a stabilizing agent.
  • a liquid carrier such as, i.e., water or a saline solution
  • lactoferrin or lactoferrin related peptides may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or inhalation.
  • topical i.e., transdermal
  • mucosal administration intranasal, vaginal, etc.
  • inhalation inhalation
  • compositions for topical administration may include the active compound formulated for a medicated application such as an ointment, paste, cream or powder.
  • Ointments include all oleaginous, adsorption, emulsion and water-solubly based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only.
  • Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin. Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram.
  • compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base.
  • Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the active ingredient and provide for a homogenous mixture.
  • Transdermal administration of the present invention may also comprise the use of a "patch".
  • the patch may supply one or more active substances at a predetermined rate and in a continuous manner over a fixed period of time.
  • the pharmaceutical compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described e.g., in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in its entirety).
  • the delivery of drugs using intranasal niicroparticle resins Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725, 871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts.
  • transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety).
  • aerosol refers to a colloidal system of finely divided solid of liquid particles dispersed in a liquefied or pressurized gas propellant.
  • the typical aerosol of the present invention for inhalation will consist of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent.
  • Suitable propellants include hydrocarbons and hydrocarbon ethers.
  • Suitable containers will vary according to the pressure requirements of the propellant.
  • Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
  • compositions of this invention can be incorporated into various types of ophthalmic formulations for delivery to the eye (e. g., topically, intracamerally, or via an implant).
  • the compositions are preferably incorporated into topical ophthalmic formulations for delivery to the eye.
  • the compositions may be combined with ophthalmologically acceptable preservatives, surfactants, viscosity enhancers, penetration enhancers, buffers, sodium chloride, and water to form an aqueous, sterile ophthalmic suspension or solution.
  • Ophthalmic solution formulations may be prepared by dissolving a compositions in a physiologically acceptable isotonic aqueous buffer.
  • the ophthalmic solution may include an ophthalmologically acceptable surfactant to assist in dissolving the compositions.
  • the ophthalmic solution may contain an agent to increase viscosity, such as, hydroxymethylcellulose, hydroxyethylcellulose, hydroxypropylmethylcellulose, methylcellulose, polyvinylpyrrolidone, or the like, to improve the retention of the formulation in the conjunctival sac.
  • Gelling agents can also be used, including, but not limited to, gellan and xanthan gum.
  • the active ingredient is combined with a preservative in an appropriate vehicle, such as, mineral oil, liquid lanolin, or white petrolatum.
  • Sterile ophthalmic gel formulations may be prepared by suspending the compositions in a hydrophilic base prepared from the combination of, for example, carbopol-974, or the like, according to the published formulations for analogous ophthalmic preparations; preservatives and tonicity agents can be incorporated.
  • composition for topical administration which is combined with a semi-solid carrier can be further formulated into a gel ointment.
  • a preferred carrier for the formation of a gel ointment is a gel polymer. Gel polymers prevent denaturation of the composition in the open skin by serum proteases.
  • the gel formulation of the present invention also provides a controlled delivery system for lactoferrin or its activity on a wound site. Controlled delivery refers to drug release or activity release sufficient to maintain a therapeutic level over an extended period of time, such as up to 24 hours or more, preferably in the range of 1 to 12 hours.
  • the present gel formulation increases the contact time of the lactoferrin at the wound site and provides a sustained release dosage form necessary to achieve a significant increase in the rate of wound healing. This is an important advantage because it permits less frequent application of the formulation to the wound and thereby permits fewer disturbances to the wound and its cellular components.
  • the gel formulation of the present invention has the advantage of adhering to a wound and conforming to irregular body or wound contours.
  • the gels may be applied directly to a wound site or in conjunction with a compliant porous or microporous substrate, for example in the form of a coating, to be applied to the wound site.
  • Gels have the further advantages of having a high water content (which keeps the wound moist), the ability to absorb wound exudate, easy application to a wound and easy removal by washing. Gels have a cool feeling when applied to a wound and thus can increase patient comfort and acceptance of the formulation, especially on sensitive wounds.
  • the aqueous gels of the present invention have different viscosities depending on the intended application of the gel.
  • Viscosity is a measure of the resistance of a liquid to flow. It is defined as the ratio of the shearing stress to the rate of shearing.
  • the shear stress is the resistance of the liquid to flow under the influence of an applied force, i.e., the molecular resistance within a body opposing an external force.
  • the shear stress is defined as the ratio of the force to the area sheared.
  • Viscosity has the dimensions of dynes/sec/cm 2 . These dimensions are referred to as poise. The dimensions of viscosity referred to herein, unless otherwise indicated, are in centipoise (cP) as measured using a Brookfield viscometer. All viscosity values are at room temperature, e.g., 22 0 C- 25°C, unless otherwise indicated.
  • the gel forming materials of the present invention may be water-soluble polymers capable of forming a viscous aqueous solution or non-water soluble, water swellable polymers (e.g., collagen), which can also form a viscous solution.
  • Swellable polymers are those that absorb water rather than dissolve in water.
  • Cross-linked forms of the polymer described herein may not be water soluble but may be water-swellable. Therefore, cross-linked forms of the polymer are within the scope of the present invention.
  • Cross-linking refers to covalently bonding polymer chains together with a bifunctional reagent such as glutaraldehyde.
  • polymers may have to be used in the salt form or partially neutralized in order to be made water soluble.
  • hyaluronic acid as sodium hyaluronate to provide suitable water solubility.
  • the polymer may be selected from the group consisting of vinyl polymers, polyoxyethylene- polyoxypropylene copolymers, polysaccharides, proteins, poly(ethylene oxide), acrylamide polymers and derivatives or salts thereof. It is understood that poly(ethyleneoxide) includes polyethylene glycol.
  • the polymers may be the same except that it is not preferred to use the polyoxyethylene- polyoxypropylene copolymers or poly(ethylene oxide).
  • the polymer is biodegradable, i.e., it will break down into harmless constituents that can be drained from or metabolized in the anterior chamber.
  • the gel forming polymers may be the same as for topical or incisional wound healing, except that poly(ethylene oxide) is not preferred to be used.
  • the vinyl polymers useful in the present invention may be selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyvinyl pyrrolidone and polyvinyl alcohol.
  • the polysaccharides useful in the present invention are selected from the group consisting of cellulose or cellulose derivatives, glycosaminoglycans, agar, pectin, alginic acid, dextran, starch, and chitosan. Starch occurs in two forms, ⁇ -amylose and amylopectin. The more water-soluble ⁇ -amylose is preferred.
  • the glycosaminoglycans are selected from the group consisting of hyaluronic acid, chondroitin, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratan sulfate, heparin sulfate and heparin.
  • the glycosaminoglycans are used to enhance wound healing in combination with any other gel-forming polymer.
  • the proteins useful in the present invention are selected from the group consisting of collagen, gelatin and fibronectin.
  • the acrylamide polymers are polyacrylamide or polymethacrylamide polymers. Biocompatible polyacrylamide polymers are preferred, hi further embodiments, carbomers are the preferred polyacrylamide polymer.
  • Carbomers are synthetic high molecular weight polymers of acrylic acid cross linked with either alkyl esters of sucrose or pentaerythritol. Suitable commercially available grades of carbomer include Carbopol 910, Carbopol 934P, Carbopol 940, Carbopol 941, Carbopol 971P, Carbopol 974P, Carbopol 980, Carbopol 981, Carbopol 1342, Rheogic 252L, Rheogic 250H, and Hostacerin PN73.
  • the viscosity may be within the range 1,000-12,000,000 cps at room temperature. It is preferred that the viscosity range be 50,000-2,000,000.
  • the topical gel formulation may comprise 0.01-5% by weight polyacrylic acid having an average molecular weight of about 450,000-4,000,000.
  • the polyacrylic acid is present at 0.5-1.5% by weight and has an average molecular weight of 2,000,000-4,000,000.
  • the pH of the polyacrylic acid gel should be within the range 4.5-8 and more preferably in the range 6.5-7.5.
  • the topical and incisional gel of the present invention may comprise 15-60% by weight of a polyoxyethylene-polyoxypropylene block copolymer having an average molecular weight of about 500-50,000.
  • the block copolymer is present at 15-40% by weight and has an average molecular weight in the range 1,000-15,000.
  • the block copolymers used in the present invention are commonly known as Pluronics.
  • Pluronics are Pluronic F88 and F 127.
  • the topical or incisional gel may comprise 1 to 20% by weight of a cellulose polymer having a molecular weight of about 50,000 to 700,000.
  • the cellulose polymer is present at 2-8% by weight and has an average molecular weight in the range 80,000-240,000.
  • Preferred cellulose polymers are hydroxypropylmethyl cellulose (HPMC), carboxymethyl cellulose (CMC) and methyl cellulose (MC).
  • the topical and incisional gel may comprise 0.5- 10% by weight of hyaluronic acid having an average molecular weight in the range 500,000 to 8,000,000.
  • the hyaluronic acid is present at 1.5-6.0% by weight and the average molecular weight is greater than 1,000,000.
  • Acrylamide polymers may be useful for all types of wound healing, particularly in the anterior chamber of the eye.
  • An absorbable acrylamide polymer such as polyacrylamide, may be a good substitute for present carrier systems used in ophthalmic applications, such as hyaluronic acid.
  • the acrylamide polymers may have an average molecular weight in the range 1-13 million, preferably about 4-6 million.
  • the weight percent of the acrylamide polymer in the gel may be 2-5%, preferably 3.5-4.5%.
  • Substituted acrylamide polymers, such as methyl and alkyl substituted polymers are also within the scope of the present invention.
  • an acrylamide gel delivery system For use in the anterior chamber of the eye, an acrylamide gel delivery system has the following characteristics: any products of the dissolution or degradation of the delivery matrix are nontoxic and do not clog the trabecular mesh work; the gel is optically transparent; and the gel can be left in the anterior chamber without causing adverse clinical effects such as an unacceptable increase in ocular pressure.
  • the desired viscosity range may be achieved by varying the molecular weight and percent concentration of the polymer in the formulation.
  • a gel having a low viscosity may be achieved by using a low molecular weight polymer or a lower percent concentration or a combination of the two.
  • a high viscosity gel may be achieved by using a higher molecular weight polymer and a higher percent concentration.
  • Intermediate viscosities may be achieved by varying the molecular weight and percent concentration accordingly.
  • the low viscosity solution may comprise 0.01-2.0% by weight polyacrylic acid having an molecular weight of about 100,000-4,000,000.
  • the polymer is present at 0.05-0.5%.
  • this dilute viscous solution may comprise 2-40% by weight of a polyoxyethylene-polyoxypropylene copolymer having an average molecular weight of 500-500,000.
  • the concentration is 2-20% and the molecular weight is 1,000-15,000.
  • the dilute viscous solution may comprise a cellulose polymer at 1-20% and having a molecular weight of about 80,000-240,000. It is preferred that the concentration be in the range of 1-10%.
  • the dilute viscous solution may comprise 0.5-5.0% by weight hyaluronic acid having an average molecular weight of about 500,000-8,000,000. Preferably, the concentration is 0.5-2.0% and the average molecular weight is 1,000,000-6,000,000. If the dilute viscous solution is to be used as eye drops, it is preferred that the viscosity be in the range 1-100 cps. If it is used for other applications, such as soaking a bandage, then any viscosity in the range 1.0-5,000 will be suitable. [0147] Upon formulation, solutions are administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective to result in an improvement or remediation of the symptoms.
  • the formulations are easily administered in a variety of dosage forms such as ingestible solutions, drug release capsules, gel ointments and the like. Some variation in dosage can occur depending on the condition of the subject being treated. The person responsible for administration can, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations meet sterility, general safety and purity standards as required by FDA Office of Biologies standards.
  • a lactoferrin composition provided in any of the above-described pharmaceutical carriers is orally, topically, or parenterally administered to a subject suspected of or having a skin, fibrotic, wound, inflammatory, autoimmune, or hyperproliferative condition.
  • One of skill in the art can determine the therapeutically effective amount of lactoferrin to be administered to a subject based upon several considerations, such as absorption, metabolism, method of delivery, age, weight, disease severity and response to the therapy.
  • Oral administration of the lactoferrin composition includes oral, buccal, enteral or intragastric administration. It is also envisioned that the composition may be used as a food additive.
  • Topical administration of the lactoferrin composition includes topical, dermal, epidermal, or subcutaneous administration.
  • Parenteral administration includes intramuscular, intravenous, intraperitoneal, and intraocular administration.
  • a further embodiment is a method of using lactoferrin to modulate the immune system through its ability to both function directly as a chemokine and to modulate other key chemokines through inducing or inhibiting chemokine production or inhibiting chemokine activity.
  • the other chemokines include but are not limited to MIP-I alpha, MIP- l ⁇ , IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha, TECK, I-TAC, Lymphotactin, PARC, MIP-ldelta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO- ⁇ , GRO- ⁇ , GRO- ⁇ , SCF, MEC/CCL28.
  • MIP-I alpha MIP- l ⁇ , IL-8
  • MIP-2 MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha
  • TECK MIP-3alpha
  • I-TAC Lymphotactin
  • PARC MIP-ldelta
  • C-TACK TARC
  • HCC-4 HCC-4
  • RANTES Eo
  • receptors to lactoferrin have not been fully characterized or identified, different tissues or cell types appear to express their own lactoferrin receptors and at least some of these may be involved in the modulation of immune, inflammatory, or growth responses [Suzuki and Lonnerdal 2002].
  • the receptors involved in this method include but are not limited to IL-8 alpha receptor, IL-8 beta receptor, neuropeptide Yl receptor, PACAP receptor, K+ channel receptor, CCR2, CCR3,, CCR6, CXCR4, EGFR, TLR-2, TLR-3, TLR-4, TLR4/MD, Mannose receptor, CD 14, TRAIL R4.
  • lactoferrin affects cell-mediated host responses.
  • LF has been shown to enhance the delayed-type hypersensitivity (DTH) response in mice, mediated by macrophages and T cells, up-regulating IL-15, MIP-lalpha, and MIP-2 (Actor 2002).
  • DTH delayed-type hypersensitivity
  • IL-15 up-regulating IL-15
  • MIP-lalpha up-regulating IL-15
  • MIP-2 up-regulating IL-15
  • MIP-2 MIP-2
  • LF also accelerates T-cell maturation by inducing the expression of the CD4 surface marker in vitro [Dhennin-Duthille 2000].
  • LF has been shown to activate natural killer NK cells, induce colony-stimulating activity, activate polymorphonuclear cells (PMNs), regulate granulopoeisis, enhance antibody-dependent cell cytotoxicity, stimulate lymphokine-activated killer cell (LAK) activity, and potentiate macrophage toxicity [Shau 1992, Horowitz 1984, Gahr 1991].
  • PMNs polymorphonuclear cells
  • LAK lymphokine-activated killer cell
  • LF can exert an anti-cancer effect.
  • Lactoferrin has a significant effect on NK cells cytotoxicity against hematopoietic and breast epithelial cell lines [Bezault 1994].
  • Research by Kuhara, et al. [Kuhara 2000] first linked the oral administration of LF, induction of IL- 18, immunomodulation, and protection against experimental cancer metastasis (lung colonization by colon 26 carcinoma).
  • LF increased CD4+, CD8+ and NK cells in peripheral blood of tumor bearing mice (P ⁇ 0.01).
  • cytotoxic activity of NK cells from white blood cells (WBC) and splenic cells obtained from mice fed LF was enhanced when tested with cultured ⁇ ac-1 and Co26Lu carcinoma cells [Kuhara, et al. 2000].
  • Oral LF has been also reported to exhibit an anti-angiogenesis activity that could be involved in inhibiting tumor growth and metastasis [Norrby 2001].
  • LF can directly mediate cell cycle arrest of cancer cells via inhibition of Gl cyclin-dependent kinases, which in addition could render such cells more vulnerable to the triggering of apoptosis [Damiens 1999].
  • LF has been shown to protect against pathogens via its immunomodulatory activity.
  • LF prolonged survival and decreased splenic viral titers in vivo in a mouse leukemia model of Friend virus complex (FVC-P) and was synergistic with IFN-gamma.
  • FVC-P Friend virus complex
  • LF and rmuIFN- gamma returned NK cell activity, decreased by FVC-P, to normal levels [Lu 1991]. Protection against a potentially lethal infection with murine Cytomegalovirus (CMV) was also observed in vivo and appeared to be mediated by an up-regulation of NK cells that was dependent on T cells [Shimizu 1996].
  • CMV Cytomegalovirus
  • LF has several anti-inflammatory properties. Previous clinical studies with lactoferrin suggest that it appears to reduce gut inflammation in patients with severe graft-vs.- host disease (GVHD) [Inoue 2001]. Th2-dominated responses are involved in the pathogenesis of GVDH and thus a reversal towards a ThI environment appears to be protective [Singh 1999, Romagnani 2000].
  • LF stimulates a shift in immune responses from Th2 to ThI by up-regulating IL-18, IFN-gamma, and down-regulating IL-4, IL-5, and IL-IO [Zhang 2001, Guillen 2002]. Consequently, this switch to a ThI response might be the mechanism by which LF mediates the anti-GVDH effect.
  • oral LF was shown to protect piglets against bacterial lipopolysaccharide (LPS) -induced sepsis [Lee 1998]. Similar protection against LPS-induced mortality as well as inhibition of key inflammatory cytokines was observed with oral lactoferrin.
  • LPS lipopolysaccharide
  • human LF has been tested in a sheep model of asthma for its ability to inhibit antigen-induced bronchoconstriction (Elrod 1997). These sheep typically develop early- and late- phase bronchoconstriction and an associated increase in bronchial responsiveness following antigen challenge. Sheep treated with lactoferrin had substantial reductions in late-phase bronchoconstriction, airway hyperresponsiveness and inflammatory cell infiltration following antigen challenge compared to untreated sheep [unpublished data].
  • the route of administration will vary, naturally, with the location and nature of the lesion, and include, for example parentally, orally or topically.
  • Parenteral administrations include, but are not limited to intradermal, transdermal, intravenous, intramuscular, intranasal, subcutaneous, percutaneous, intratracheal, intraocular, intraperitoneal, intratumoral, perfusion, lavage, direct injection U.S. Pat. Nos. 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363 (each specifically incorporated herein by reference in its entirety).
  • Alimentary administrations include, but are not limited to orally, buccally, rectally, or sublingually. W
  • Treatment methods will involve treating an individual with an effective amount of a lactoferrin composition as defined in the present invention.
  • An effective amount is described, generally, as that amount sufficient to detectably and repeatedly to ameliorate, reduce, minimize or limit the extent of a disease or its symptoms.
  • the effective amount or "therapeutically effective amounts" of the lactoferrin composition to be used are those amounts effective to produce beneficial results, particularly with respect to cancer treatment, in the recipient animal or patient. Such amounts may be initially determined by reviewing the published literature, by conducting in vitro tests or by conducting metabolic studies in healthy experimental animals. Before use in a clinical setting, it may be beneficial to conduct confirmatory studies in an animal model, preferably a widely accepted animal model of the particular disease to be treated. Preferred animal models for use in certain embodiments are rodent models, which are preferred because they are economical to use and, particularly, because the results gained are widely accepted as predictive of clinical value.
  • a specific dose level of active compounds such as the lactoferrin composition for any particular patient depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy. The person responsible for administration will determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologies standards.
  • a therapeutically effective amount of a lactoferrin composition as a treatment varies depending upon the host treated and the particular mode of administration.
  • the dose range of a lactoferrin composition will be about 0.5 mg/kg body weight to about 500 mg/kg body weight.
  • body weight is applicable when an animal is being treated. When isolated cells are being treated, “body weight” as used herein should read to mean “total cell body weight”. The term “total body weight” may be used to apply to both isolated cell and animal treatment. AU concentrations and treatment levels are expressed as “body weight” or simply “kg” in this application are also considered to cover the analogous "total cell body weight "and" total body weight” concentrations.
  • the treatments may include various "unit doses.”
  • Unit dose is defined as containing a predetermined quantity of the therapeutic composition calculated to produce the desired responses in association with its administration, i.e., the appropriate route and treatment regimen.
  • the quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts.
  • Also of import is the subject to be treated, in particular, the state of the subject and the protection desired.
  • a unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time.
  • the composition is given in a single dose or multiple doses.
  • the single dose may be administered daily, or multiple times a day, or multiple times a week, or monthly or multiple times a month.
  • the composition is given in a series of doses. The series of doses may be administered daily, or multiple times a day, weekly, or multiple times a week, or monthly, or multiple times a month.
  • the hyperproliferative disease includes but is not limited to neoplasms.
  • a neoplasm is an abnormal tissue growth, generally forming a distinct mass that grows by cellular proliferation more rapidly than normal tissue growth.
  • Neoplasms show partial or total lack of structural organization and functional coordination with normal tissue. These can be broadly classified into three major types. Malignant neoplasms arising from epithelial structures are called carcinomas, malignant neoplasms that originate from connective tissues such as muscle, cartilage, fat or bone are called sarcomas and malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) including components of the immune system, are called leukemias, lymphomas and myelomas.
  • a tumor is the neoplastic growth of the disease cancer.
  • a "neoplasm”, also referred to as a “tumor” is intended to encompass hematopoietic neoplasms as well as solid neoplasms.
  • neoplasms include, but are not limited to melanoma, non-small cell lung, small-cell lung, lung, hepatocarcinoma, retinoblastoma, astrocytoma, gliobastoma, gum, tongue, leukemia, neuroblastoma, head, neck, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, sarcoma, cervical, gastrointestinal, lymphoma, brain, colon, bladder, myeloma, or other malignant or benign neoplasms.
  • hyperproliferative diseases include, but are not limited to neurofibromatosis, rheumatoid arthritis, Waginer's granulomatosis, Kawasaki's disease, lupus erathematosis, midline granuloma, inflammatory bowel disease, osteoarthritis, leiomyomas, adenomas, lipomas, hemangiomas, fibromas, vascular occlusion, restenosis, atherosclerosis, preneoplastic lesions, carcinoma in situ, oral hairy leukoplakia, or psoriasis, and pre-leukemias, anemia with excess blasts, and myelodysplastic syndrome.
  • neoplasms of interest in the present invention include, but are not limited to hematopoietic neoplasms.
  • a hematopoietic neoplasm may include acute myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, chronic lymphocytic leukemia or other malignancy of hematologic origin.
  • the lactoferrin compositions are administered in an effective amount to decrease, reduce, inhibit or abrogate the growth of a tumor.
  • the amount may vary from about 0.1 ⁇ g to about lOOg of the lactoferrin composition.
  • the lactoferrin composition is orally administered in the range of lmg to lOOg per day, more preferably about 20mg to about 1Og per day with the most preferred dose being 4.5g per day.
  • Intravenously administered lactoferrin can be in the range of 0.1 ⁇ g to about to 1Og per day, more preferably about 0.1 ⁇ g to about lmg with the most preferred dose being 250 mg per day.
  • a lactoferrin composition is intratumorally administered in the range of 0.1 ⁇ g to 1Og per day with the most preferred dose being lOO ⁇ g per day.
  • the amount of the lactoferrin composition may vary from about l ⁇ g to about 10Og.
  • the topical gel, solution, capsule or tablet comprises a concentration of about 0.01% to about 20% of lactoferrin related peptides and/or lactoferrin N-terminal variants. More preferably, the topical gel, solution, capsule or tablet may comprise a concentration of about 1% to about 8.5% lactoferrin related peptides and/or lactoferrin N-terminal variants.
  • intratumoral administration of the composition includes intratumoral injection, electroporation, or surgical or endoscopic implantation.
  • Intratumoral injection, or injection into the tumor vasculature is specifically contemplated for discrete, solid, accessible lumorsT Local, regional or systemic administration also may be appropriate.
  • the tumor being treated may not, at least initially, be resectable.
  • Treatments with the lactoferrin composition may increase the resectability of the tumor due to shrinkage at the margins or by elimination of certain particularly invasive portions. Following treatments, resection may be possible. Additional treatments subsequent to resection will serve to eliminate microscopic residual disease at the tumor site.
  • the present invention may be used at the time of surgery, and/or thereafter, to treat residual or metastatic disease.
  • a resected tumor bed may be injected or perfused with a formulation comprising the lactoferrin composition.
  • the perfusion may be continued post-resection, for example, by leaving a catheter implanted at the site of the surgery.
  • Periodic post-surgical treatment is also envisioned.
  • Continuous administration also may be applied where appropriate, for example, where a tumor is excised and the tumor bed is treated to eliminate residual, microscopic disease. Delivery via syringe or catherization is preferred.
  • Such continuous perfusion may take place for a period from about 1-2 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 weeks or longer following the initiation of treatment.
  • the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs.
  • limb perfusion may be used to administer therapeutic compositions of the present invention, particularly in the treatment of melanomas and sarcomas.
  • a further embodiment is a method of enhancing a mucosal immune response in the gastrointestinal tract in a subject comprising the step of administering orally to said subject a lactoferrin composition. It is also envisioned that the lactoferrin composition stimulates interleukin-18 following oral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
  • a further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing the systemic immune system " by increasing the amount of the lactoferrin composition in the systemic circulation.
  • the composition is administered intravenously. It is envisioned that the lactoferrin composition stimulate interleukin-18 following intravenous administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
  • a further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing a local or systemic immune system by increasing the amount of lactoferrin in the vicinity of the tumor.
  • Vicinity of the tumor refers to the general area of the tumor, for example the lactoferrin can be administered directly into or on the tumor, or in the general area of the tumor, but not directly into the tumor.
  • the general area may include the margin area or near or adjacent the margin area of the tumor.
  • the lactoferrin composition is administered intratumorally. It is envisioned that the lactoferrin composition stimulates interleukin-18 following intratumoral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
  • a further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing a local or systemic immune system by increasing the amount of a lactoferrin composition in the skin in the vicinity of the tumor.
  • the composition is administered topically.
  • administration in the vicinity of the tumor includes administration near or adjacent to the margins of the tumor or directly in the margin area of the tumor.
  • the lactoferrin composition stimulates interleukin-18 and GM-CSF in the local tissue (e.g., keratinocytes), which enhances immune cells.
  • the lactoferrin composition stimulate interleukin-18 following intratumoral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
  • an "anti-cancer” agent is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, or increasing the lifespan of a subject with cancer.
  • Anti-cancer agents include biological agents (biotherapy), chemotherapy agents, and radiotherapy agents.
  • these other compositions would be provided in a combined amount effective to kill or inhibit proliferation of the cell.
  • This process may involve administering the lactoferrin composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s).
  • the lactoferrin composition of the present invention may precede or follow the other anti-cancer agent treatment by intervals ranging from minutes to weeks.
  • the other anti-cancer agent and lactoferrin composition are administered or applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and lactoferrin composition would still be able to exert an advantageously combined effect on the cell.
  • Cancer therapies also include a variety of chemical based treatments.
  • Some examples of chemotherapeutic agents include without limitation antibiotic chemotherapeutics such as Doxorubicin, Daunorubicin, Adriamycin, Mitomycin (also known as mutamycin and/or mitomycin-C), Actinomycin D (Dactinomycin), Bleomycin, Plicomycin, plant alkaloids such as Taxol, Vincristine, Vinblastine, miscellaneous agents such as platinum based agents (e.g., Cisplatin (CDDP)), etoposide (VP 16), Tumor Necrosis Factor, and alkylating agents such as, Carmustine, Melphalan (also known as alkeran, L-phenylalanine mustard, phenylalanine mustard, L-PAM, or L-sarcolysin, (a phenylalanine derivative of nitrogen mustard), Cyclophosphamide, Chlorambucil, Busulfan (also known as myleran), tax
  • Some examples of other agents include, but are not limited to, Carboplatin, Procarbazine, Mechlorethamine, Irinotecan, Topotecan, Ifosfamide, Nitrosurea, Etoposide (VP 16), Tamoxifen, Raloxifene, Toremifene, Idoxifene, Droloxifene, TAT-59, Zindoxifene, Trioxifene, ICI 182,780, EM-800, Estrogen Receptor Binding Agents, Gemcitabinen, Navelbine, Farnesyl-protein transferase inhibitors, Transplatinum, 5-Fluorouracil, hydrogen peroxide, and Methotrexate, Temazolomide (an aqueous form of DTIC), Mylotarg, Dolastatin-10, Bryostatin, or any analog or derivative variant of the foregoing.
  • Radiotherapeutic Agents include, but are not limited to, Carboplatin, Procarbazine, Mech
  • Radiotherapeutic agents and factors include radiation and waves that induce DNA damage for example, ⁇ -irradiation, X-rays, UV-irradiation, microwaves, electronic emissions, radioisotopes, and the like. Therapy may be achieved by irradiating the localized tumor site with the above described forms of radiations. It is most likely that all of these factors effect a broad range of damage to DNA 5 the precursors of DNA, the replication and repair of DNA, and the assembly and maintenance of chromosomes.
  • Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 weeks), to single doses of 2000 to 6000 roentgens.
  • Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
  • Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene therapy, immunotherapy and/or alternative therapies.
  • Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed.
  • Tumor resection refers to physical removal of at least part of a tumor.
  • treatment by surgery includes laser surgery, cryosurgery, electrosurgery, and miscopically controlled surgery (Mohs' surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue.
  • a cavity may be formed in the body.
  • Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy. Such treatment may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. These treatments may be of varying dosages as well.
  • Other Biotherapy Agents include:
  • additional agents include, without limitation, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adhesion, agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers, or other biological agents, as well as biotherapy such as for example, hyperthermia.
  • Hyperthermia is a procedure in which a patient's tissue is exposed to high temperatures (up to 106°F). External or internal heating devices may be involved in the application of local, regional, or whole-body hyperthermia. Local hyperthermia involves the application of heat to a small area, such as a tumor. Heat may be generated externally with high- frequency waves targeting a tumor from a device outside the body. Internal heat may involve a sterile probe, including thin, heated wires or hollow tubes filled with warm water, implanted microwave antennae, or radiofrequency electrodes.
  • a patient's organ or a limb is heated for regional therapy, which is accomplished using devices that produce high energy, such as magnets.
  • some of the patient's blood may be removed and heated before being perfused into an area that will be internally heated.
  • Whole-body heating may also be implemented in cases where cancer has spread throughout the body. Warm-water blankets, hot wax, inductive coils, and thermal chambers may be used for this purpose.
  • Hormonal therapy may also be used in conjunction with the present invention.
  • the use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or estrogen and this often reduces the risk of metastases.
  • Adjuvant therapy may also be used in conjunction with the present invention.
  • adjuvants or immunomodulatory agents include, but are not limited to tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-I, MlP-lbeta, MCP-I, RANTES, and other chemokines.
  • Immunotherapy includes, but are not limited to tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-I, MlP-lbeta, MCP-I, RANTES, and other chemokines.
  • Immunotherapeutics generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells.
  • the immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell.
  • the antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing.
  • the antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent.
  • the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target.
  • Various effector cells include cytotoxic T cells and NK cells.
  • vaccines that are used to treat cancer may be used in combination with the present invention to improve the therapeutic efficacy of the treatment.
  • Such vaccines include peptide vaccines or dendritic cell vaccines.
  • Peptide vaccines may include any tumor-specific antigen that is recognized by cytolytic T lymphocytes.
  • dendritic cell vaccination comprises dendritic cells that are pulsed with a peptide or antigen and the pulsed dendritic cells are administered to the patient.
  • tumor-specific antigens that are being used as vaccines in melanoma include, but are not limited to gplOO or MAGE-3. These antigens are -being - administered as peptide vaccines and/or as dendritic cell vaccines.
  • the present invention is designed for the treatment of any type of skin disease including but not limited to psoriasis, scleroderma, atopic dermatitis, epidermolysis bullosa, acute wounds, chronic wounds, and burns.
  • the present invention is designed for the treatment of any type of wound, which includes, but is not limited to skin wound, internal wound, gastrointestinal wound, oral wound, bone wounds, ophthalmic wound, surgical wound, or any combination thereof.
  • Wounds can be found on but not limited to skin, internal organs, stomach and intestines (gastrointestinal), oral mucosa, and eye (ophthalmic wounds, e.g., corneal ulcers, radiokeratotomy, corneal transplants, epikeratophakia and other surgically induced wounds in the eye).
  • wounds can also be classified as but are not limited to incisional wounds, excisional wounds, diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers, chemical wounds, burn wounds, full-thickness skin wound and partial-thickness skin wound.
  • a further embodiment of the present invention is administering the inventive composition to treat skin wounds.
  • Skin wounds further comprise but are not limited to full-thickness wounds and partial-thickness wounds.
  • Full-thickness wounds involve the complete removal of epidermis and dermis to the depth of fascial planes or subcutaneous fat.
  • the thin musculature of the panniculus carnosus which firmly adheres to the base of the dermis, is usually removed as well.
  • partial-thickness wounds a substantial amount of dermis, mostly reticular, is left behind, and, more importantly, the bases of most epidermal appendages (sebaceous and sweat glands, hair follicles) remain intact.
  • a wound can be further defined as an acute wound.
  • Acute wounds have a relatively rapid rate of healing, especially in healthy subjects. However, in the elderly or immunocompromised healing can be prolonged. Healing is also prolonged if the wound becomes infected.
  • Preferred acute wounds that are to be treated with the present composition include, but are not limited to partial-thickness burns, lacerations, bullet wounds or infected wounds.
  • a wound is also further defined as a chronic wound.
  • chronic wounds or chronic ulcers include, but are not limited to diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers.
  • chronic wounds can also include infected wounds.
  • Chronic wounds are wounds that do not repair or do so extremely slowly, and show partial or total lack of structural organization and functional coordination with normal tissue.
  • Chronic wounds or chronic ulcers can be broadly classified into three major types: diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers. Diabetic ulcers often occur on a foot.
  • venous ulcers venous hypertension causes disturbed microcirculation and pathological changes of the capillaries, elevated persistent levels of pro-inflammatory cytokines and proteases. Fibroblast senesce and respond less to growth factors, which distribute unfavorably. Proteolytic enzymes and their inhibitors are imbalanced. Pressure ulcers occur when skin is under pressure without movement to allow blood flow for 8-12 hours.
  • the inventive composition is administered in an effective amount to seal, to close, to improve or to repair the wound.
  • the composition of the present invention can also decrease, reduce, or inhibit, bacterial infections of the wound, which aid in the healing process of a wound.
  • the lactoferrin composition of the present invention may be administered for any given period of time until the wound is healed at least by 5%, 10%, 20%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% or any range in between.
  • the gel formulation of the present invention may be used to coat fibers of an absorbent gauze dressing to form a wound healing bandage which may then be placed on a wound.
  • the low viscosity formulation is preferred for this use.
  • the wound healing bandage may be prepared by soaking a gauze dressing with an aqueous gel solution containing lactoferrin having wound healing activity. The bandage can then be applied to the wound scTthat the coated fibers of the gauze contacts the wound and stimulate the rate of wound healing.
  • the gel forming polymer be biodegradable.
  • the naturally occurring polymers are generally biodegradable. Examples of these are collagen, the glycosaminoglycans, gelatin and starch.
  • the cellulosics are not biodegradable.
  • the synthetic polymers such as the vinyl polymers are not degradable.
  • the biodegradability of the polymers described herein is well known to those skilled in the art.
  • a further embodiment of the present invention is a method of treating a wound comprising the step of supplementing the local immune system by increasing the amount of a lactoferrin composition in the vicinity of the wound.
  • the peptide is administered topically to the wound.
  • the present invention also provides a method of treating a wound comprising the step of supplementing the systemic immune system by increasing the amount of a lactoferrin composition in the systemic circulation.
  • the lactoferrin is administered via a parenteral route, which includes, but is not limited to intramuscular, intravenous, intraperitoneal, intraoccular, intraarticular or into a surgical field.
  • the present invention provides a method of enhancing the immune system of a subject suffering from a wound by administering to the subject a lactoferrin composition.
  • a lactoferrin composition Depending upon the mode of administration, different arms of the immune system are enhanced.
  • topical administration of the composition results in enhancement of the local immune system, i.e., in the vicinity of the wound.
  • Parenteral administration of the composition results in enhancement of the systemic immune system.
  • oral administration of the composition results in enhancement of the mucosal immune system, which can also result in systemic effects as well.
  • IL- 18 or GM-CSF stimulate the production or activity of cells involved in wound repair, for example, but not limited to keratinocytes, endothelial cells, dendritic cells, fibroblasts, and myofibroblasts.
  • the lactoferrin composition inhibit the production of TNF-alpha, which may lead to excess inflammation and tissue destruction for example by stimulating the production of metalloproteinases.
  • compositions comprising a lactoferrin peptide related compound may stimulate the production of a cytokine or a chemokine.
  • Cytokines for example, interleukin-18 or granulocyte/macrophage colony-stimulating factor, can also stimulate the production or activity of cells involved in wound repair.
  • the cells involved in wound repair include, but are not limited to keratinocytes, endothelial cells, fibroblasts, dendritic cells, and myofibroblasts.
  • the inhibition of TNF-alpha further inhibits the migration and maturation of dendritic cells and the production of metalloproteinases.
  • the dendritic cells can be Langerhans cells.
  • wound healing agents are capable of negatively affecting a wound in a subject, for example, by enhancing the growth rate of skin cells, augmenting the blood supply to skin cells, promoting an immune response against bacteria infecting the wound, killing bacteria, cleaning ischemic tissue, promoting the closure of the wound.
  • these other wound healing agents are provided in a combined amount effective to promote the healing of a wound.
  • This process may involve administering the composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s).
  • the composition of the present invention may precede or follow the other wound healing agent treatment by intervals ranging from minutes to weeks.
  • the other wound healing agent and inventive composition are administered or applied separately to the wound, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and human lactoferrin composition would still be able to exert an advantageously combined effect on the wound.
  • Wound healing therapies include growth factor based treatments. Examples include, but are not limited to RegranexTM (Becaplermin-BB gel), AuToIo-GeI (autologous activated platelet releasate), Procuren (autologous thrombin-induced platelet releasate). Growth factors act without limitation by promoting granulation or the formation of new highly vascularized connective tissue; stimulating proliferation, differentiation and migration of epithelial cells, vascular endothelial cells and other skin cells; enhancing the production of collagen, collagenase, and extracellular matrix.
  • RegranexTM Becaplermin-BB gel
  • AuToIo-GeI autologous activated platelet releasate
  • Procuren autologous thrombin-induced platelet releasate.
  • Growth factors act without limitation by promoting granulation or the formation of new highly vascularized connective tissue; stimulating proliferation, differentiation and migration of epithelial cells, vascular endothelial cells and other skin cells
  • Examples include but are not limited to Apligraf (bilayered living skin), Trancyte (Human fibroblast-derived temporary skin substitute), Dermagraft (permanent, one- layer skin substitute), Epicel (living one-layer artificial skin), Integra (collagen-based skin regeneration template), AlloDerm (single-layer artificial skin made from human cadavers), CCS (living, cultured, artificial skin).
  • Debridement is a process or procedure to clean ischemic or dead tissue.
  • Enzymatic debriders include Accuzyme papain-urea debriding ointment and Collagenase Santyl.
  • Surgical debridement refers to physical removal of at least part of the ischemic or dead tissue in a wound. Debridement may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months.
  • Enzymatic debridement treatments may be of varying dosages as well. It is further contemplated that the present invention may be used in conjunction with enzymatic or surgical debridement.
  • Wound healing therapies include a variety of treatments based on dressings.
  • Dressing categories include but are not limited to amorphous hydrogels, hydrogel sheets, absorptives, alginates, biological and synthetic dressings, collagens, composites, contact layers, elastic gauzes, foams, gauzes and non-woven dressings, hydrocolloids, impregnated dressings, silicone gel sheets, silver dressings, transparent films, wound fillers
  • Examples include but are not limited to Biolex, Lamin, Wound Wash Saline, Techni-Care, CarraKlenz, DiaB Klenz, MicroKlenz, RadiaCare Klenz, UltraKlenz, Comfee Sea-Clens, Optipore Sponge, Saf-Clens, Shur-Clens, Dermagran, DermaKlenz, Dumex, Gene Klenz, GRX, Allclenz, Restore, Hyperion, Medi Tech, Skintegrity, MPM Antimicrobial, Clins Wound, Septicare, Lobana Saline. f) Antimicrobials
  • Examples of include but are not limited to Sulfamylon Cream, Thermazene Cream (1% silver sulfadiazine), cadexomer-iodine pads or gel.
  • Examples of intravenous antimicrobials include but are not limited to imipenem/cilastatin, ⁇ -lactam/ ⁇ -lactamase inhibitors (ampicillin/sulbactam, piperacillin/tazobactam), and broad-spectrum cephalosporins (cefoxitin, ceftizoxime, ceftazidime).
  • Dynamic compression examples include pumps and sleeves such as but not limited to ArtAssist, ArterialFlow, EdemaFlow, PulStar, Circulator Boot, Flowplus, Flowpress, Flowtron.
  • Static compression include but are not limited to leg wrappings, gloves, socks, leg wears, leg supports, arm sleeves, stasis pads, compression hosieries, non-elastic bands, high compression bandages, zinc impregnated bandages, elastic bandages.
  • Examples of systemic hyperbaric oxygen therapy include but are not limited to compartments for one patient to lay down, for one patient to sit up to 25 degree angle, for one patient to sit up to 90 degree angle, for more than one patient to be treated simultaneously.
  • Examples of topical hyperbaric oxygen therapy include but are not limited to disposable topical hyperbaric oxygen systems for extremity ulcers, disposable topical hyperbaric oxygen systems for decubitis, post-op and trauma wounds.
  • Examples include but are not limited to dry hydrotherapy machines; non- contact thermal wound care systems for use on partial- and full-thickness wounds that maintain warmth and humidity in the wound area; systems that provide non-thermal, pulsed high frequency, high peak power, electromagnetic energy to treat edema and pain in acute and chronic wounds; systems that use controlled, localized negative pressure and support for moist wound healing; pulsatile irrigators with controllable pressures below 15 psi for site-specific treatment of various wounds with variety of tips; various wound irrigation and whirlpool systems.
  • Examples include but are not limited to isotonic, high-protein, fiber- containing tube feedings to support wound healing; high-protein, cholesterol-free nutritional supplements.
  • Examples include but are not limited to Dermabond, CoStasis, CoSeal, BioGlue, FibRx, FocalSeal, FloSeal, AutoSeal, Indermil, Syvek, LiquiSheild, LiquiBand, Quixil, CryoSeal, VIGuard Fibrin Sealant, and various tapes, closures, and securement products.
  • Examples include but are not limited to topical aerosols which stimulate the capillary bed of chronic wounds; skin protectants with zinc-nutrient formulations; topical gels to help scars feel softer and smoother; hydrophilic ointments that cleanse degraded proteins, promote healthy granulation, control local inflammation and reduce wound odors; oil-and- water wound dressing emulsions that selectively recruit macrophages.
  • Adjuvant therapy may also be used in conjunction with the present invention.
  • adjuvants or immunomodulatory agents include, but are not limited to tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-I, MIP-I beta, MCP-I, RANTES, and other chemokines.
  • a lactoferrin composition provided in any of the above-described pharmaceutical carriers is orally, topically, or parenterally administered to a subject suspected of or having psoriasis.
  • Psoriasis is a chronic inflammatory skin disorder that undergoes repeated relapse and remission and affects one to three percent of the world's population. The disease can begin at any age and has its peak appearance in the third decade. Its severity, course, and remissions are unpredictable. (See, T. P. Habif, in: Clinical Dermatology, Mosby, Mo. (1996) and The Textbook of Medicine edts: J. B. Wyngaarden and L. H. Smith, W., W. B. Saundres Company, Philadelphia (1985)).
  • Patients with psoriasis can be divided into two groups, those with a genetic predisposition and those that respond to epigenetic, i.e. environmental factors.
  • the most common form is chronic plaque psoriasis and is characterized by hyperplasia of the epidermis and inflammation of the dermis and epidermis. These changes arise due to activation of a T- lymphocyte cell mediated dermal immune reaction in regional lymph nodes in response to unidentified antigenic stimuli.
  • the activated T-cells cause keratinocyte cells to proliferate and assume a psoriasis phenotype.
  • the inflammatory reaction is caused by proinflammatory cytokine proteins that are induced in response to the environmental stimulus and results in infiltration of the dermis and epidermis by inflammatory white blood cells.
  • the present invention provides a method of enhancing the immune system of a subject suffering from psoriasis by administering to the subject a lactoferrin composition.
  • a lactoferrin composition Depending upon the mode of administration, different arms of the immune system are enhanced. or example, topical administration of the composition resulfs in " enhancement of the local immune system, i.e., in the vicinity of the wound.
  • Parenteral administration of the composition results in enhancement of the systemic immune system.
  • oral administration of the composition results in enhancement of the mucosal immune system, which can also result in systemic effects as well.
  • the present invention provides compositions that directly interfere with the production of TNF- ⁇ by keratinocytes, thus preventing the inappropriate local immune response causing psoriasis. More specifically, the present invention provides novel pharmaceutical compositions comprising a lactoferrin composition for the treatment of psoriasis, hi a preferred embodiment, the compositions are formulated for topical application in about 0.5% to about 5% carrier.
  • the compositions are formulated for intradermal injections.
  • This process may involve administering the composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s).
  • composition of the present invention may precede or follow the other anti-inflammatory agent treatment by intervals ranging from minutes to weeks.
  • the other anti-inflammatory agent and inventive composition are administered or applied separately to the anti-inflammatory, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and the inventive composition would still be able to exert an advantageously combined effect on the psoriasis.
  • Typical anti-inflammatory agents are topical agents, for example, but not limited to are corticosteroids (hydrocortisone and analogs).
  • corticosteroids hydrocortisone and analogs.
  • Systemic corticosteroids for example, can induce prompt resolution of psoriatic lesions, but suppression requires ever- increasing doses.
  • the composition provided in any of the above-described pharmaceutical carriers is orally administered to a subject suspected of or having a respiratory disorder.
  • the respiratory disorder can be an allergic or non-allergic respiratory disorder.
  • an allergic or non-allergic respiratory disorder includes asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis and allergic rhinitis.
  • the respiratory disorder is characterized by increased responsiveness of the trachea and bronchi to various stimuli, i.e., allergens, resulting in widespread narrowing of the airways.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the obstruction of the airway or the hyperresponsiveness of the respiratory system to the allergen or other stimuli.
  • the amount of lactoferrin composition may vary from about 1 mg to about 100 g.
  • the composition that is orally administered contains the range of 1 mg to 10 g of lactoferrin composition per day, more preferably, 10 mg to about 1 g.
  • the composition of the present invention also contains metal chelators.
  • a preferred metal chelator is EDTA.
  • the composition that is orally administered contains the ratio of 1:10,000 to about 2:1 EDTA to lactoferrin.
  • a further embodiment is a method of enhancing a mucosal immune response in the gastrointestinal tract in a subject comprising the step of administering orally to said subject the composition of the present invention. It is envisioned that oral administration of lactoferrin composition stimulates IL- 18 in the lungs, sinuses or systemically. Yet further, it is contemplated that an enhancement of lactoferrin composition can reduce the infiltration of inflammatory cells into the lung.
  • asthma agents are used in combination with the composition of the present invention.
  • agents known to treat asthma include mast cell degranulation agents (i.e., Cromylyn sodium or Nedocromil sodium), leukotriene inhibitors (i.e., Monteleukast sodium, Zafirlukast, or Pranlukast hydrate), corticosteroids (i.e., Beclomethasone, Budesonide, Ciclesonide, Hydrolysable glucocorticoid, Triamcinolone acetonide, Flunisolide, Mometasone furoate, or Fluticasone propionate), ⁇ -Antagonists (i.e., Albuterol, Bambuterol, Formoterol, Salbutamol, Terbutaline sulfate, or Salmeterol), IgE binding inhibitors (
  • the agent is a known agent used to treat allergic rhinitis.
  • exemplary agents include, but are not limited to Hl antihistamines i.e., terfendine or astemizole; alpha-adrenergic agents; and glucocorticoids, i.e., beclamethasone or flunisolide.
  • the agent is a known agent to treat sinusitis, more specifically, chronic sinusitis.
  • exemplary agents/therpies include, but are not limited to surgery (e.g., enlarging a sinus passage, remove obstructing bone or nasal polyps, mucosal stripping, removal of sinuses); corticosteroids (e.g., oral, intranasal, nebulized, or inhaled); antibiotics (e.g., oral, intranasal, nebulized, inhaled or intravenous); anti-fungal agents; salt-water nasal washes and mist sprays; anti-inflammatory agents; decongestants (oral or nasal); guaifenesin; potassium iodide; leukotriene inhibitors (e.g., monteleukast); mast cell degranulating agents; topical moisterizing applications (e.g., nasal sprays or gels which may contain moisterizing agents such as propylene glycol
  • the agent is a known agent to treat COPD or chronic bronchitis or emphysema.
  • agents/interventions include, but are not limited to oxygen; brbnchodilator drugs [e.g., short and long acting beta-2 stimulants, anticholinergics (e.g., ipratoprium bromide, theophylline compounds or a combination), steroids (topical or oral), or mucolytic agents (e.g., ambroxol, ergosterin, carbocysteine, iodinated glycerol)]; antibiotics; anti-fungals; moisterization by nebulization; anti-tussives; respiratory stimulants (e.g., doxapram, almitrine bismesylate); surgery (e.g., bullectomy, lung volume reduction surgery, lung transplantation); and alpha 1 antitrypsin administration.
  • brbnchodilator drugs e.g., short and long acting beta-2 stimulants, anticholinergics
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • combination regimens of the composition and one or more agents are employed.
  • One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination.
  • the present invention is useful for the treatment and/or prevention of any inflamatory condition that is caused by an infectious disease.
  • infectious diseases include infections of viral etiology such as HIV, West Nile Virus, influenza, Herpes, viral hepatitis, Epstein Bar, polio, viral encephalitis, measles, chicken pox, Papilloma virus etc.; or infections of bacterial etiology such as bacterial meningitis, paramenigeal infections, septic thrombophlebitis, pneumonia, tuberculosis, myocarditis, bacteremia, syphilis, etc.; or infections of parasitic etiology such as malaria, trypanosomiasis, leishmaniasis, candidiasis, trichomoniasis, amoebiasis, etc.
  • viral etiology such as HIV, West Nile Virus, influenza, Herpes, viral hepatitis, Epstein Bar, polio, viral encephalitis
  • infectious inflammatory conditions include but are not limited to pathogens such as meningococci, Salmonella typhi, Dengue virus, Plasmodia, Pneumocystis carinii, Yersinia enterocolitica pathogens, enterovirus, fungi.
  • pathogens such as meningococci, Salmonella typhi, Dengue virus, Plasmodia, Pneumocystis carinii, Yersinia enterocolitica pathogens, enterovirus, fungi.
  • the present invention can have applications therefore in the prevention and treatment of viral diseases.
  • pathogenic viruses which are mentioned by way of example, influenza A, B and C, parainfluenza, paramyxoviruses, Newcastle disease virus, respiratory syncytial virus, measles, mumps, adenoviruses, adenoassociated viruses, parvoviruses, Epstein-Barr virus (EBV), rhinoviruses, coxsackieviruses, echoviruses, reoviruses, rhabdoviruses, lymphocytic choriomeningitis, coronavirus, polioviruses, herpes simplex (HSV), human immunodeficiency viruses (HIV), cytomegaloviruses, papillomaviruses, human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), varicella-zoster, poxviruses, rub
  • the present invention is also useful in the prevention, inhibition, or treatment of bacterial infections.
  • the following bacteria are mention by way of example, including, but not limited to, serotypes of pneumococci, streptococci such as S. pyogenes, S. agalactiae, S. equi, S. canis, S. bovis, S. equinus, S. anginosus, S. sanguis, S. salivarius, S. mitis, S.
  • mutans other viridans streptococci, peptostreptococci, other related species of streptococci, enterococci such as Enterococcus faecalis, Enterococcus faecium, Staphylococci, such as Staphylococcus epidermidis, Staphylococcus aureus, particularly in the nasopharynx, Hemophilus influenzae, pseudomonas species such as Pseudomonas aeruginosa, Pseudomonas pseudomallei, Pseudomonas mallei, brucellas such as Brucella melitensis, Brucella suis, Brucella abortus, Bordetella pertussis, Neisseria meningitidis, Neisseria gonorrhoeae, Moraxella catarrhalis, Corynebacterium diphtheriae, Corynebacterium ulcerans, Coryne
  • the invention may also be useful against gram negative bacteria such as Klebsiella pneumoniae, Escherichia coli, Proteus, Serratia species, Acinetobacter, Yersinia pestis, Francisella tularensis, Enterobacter species, Bacteriodes and Legionella species and the like.
  • fungal and other mycotic pathogens or cells infected with fungal or other mycotic pathogens may be used in the present invention to prevent and/or treat diseases, rariging from mycoses involving skin, hair, or mucous membranes, such as, but not limited to, Aspergillosis, Black piedra, Candidiasis, Chromomycosis, Cryptococcosis, Onychomycosis, or Otitis externa (otomycosis), Phaeohyphomycosis, Phycomycosis, Pityriasis versicolor, ringworm, Tinea barbae, Tinea capitis, Tinea corporis, Tinea cruris, Tinea favosa, Tinea imbricata, Tinea manuum, Tinea nigra (palmaris), Tinea pedis, Tinea unguium, Torulopsosis, Trichomycos
  • Fungal and mycotic pathogens that can be used in the present invention include, but are not limited to, Absidia spp., Actinomadura madurae, Actinomyces spp., Allescheria boydii, Alternaria spp., Anthopsis deltoidea, Apophysomyces elegans, Arnium leoporinum, Aspergillus spp., Aureobasidium pullulans, Basidiobolus ranarum, Bipolaris spp., Blastomyces dermatitidis, Candida spp., Cephalosporium spp., Chaetoconidium spp., Chaetomium spp., Cladosporium spp., Coccidioides immitis, Conidiobolus spp., Corynebacterium tenuis, Cryptococcus spp., Cunninghamella bertholletiae, Curvularia spp., Dactylaria spp
  • the invention may prove useful in controlling protozoan or macroscopic infections by organisms such as Cryptosporidium, Isospora belli, Toxoplasma gondii, Trichomonas vaginalis, Cyclospora species, for example, and for Chlamydia trachomatis and other Chlamydia infections such as Chlamydia psittaci, or Chlamydia pneumoniae, for example.
  • organisms such as Cryptosporidium, Isospora belli, Toxoplasma gondii, Trichomonas vaginalis, Cyclospora species, for example, and for Chlamydia trachomatis and other Chlamydia infections such as Chlamydia psittaci, or Chlamydia pneumoniae, for example.
  • composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having bacteremia, sepsis, septic shock or sequelae.
  • bacteremia sepsis, septic shock or sequelae.
  • These conditions could be caused by gram- negative, gram-positive bacteria or other infectious agents such as Candida in any foci of the body and are at a risk of developing into or have developed into a systemic inflammatory response syndrome.
  • bacteremia may be caused by surgical manipulation of infected oral tissues or routine dental manipulations; catheterization of an infected lower urinary tract; incision and drainage of an abscess; and colonization of indwelling devices, especially IV and intracardiac catheters, urethral catheters, and ostomy devices and tubes.
  • the primary site of infection is usually in the lungs, in the GU or GI tract, or in soft tissues including the skin in patients with decubitus ulcer.
  • gram- negative bacteremia occurs more commonly, than in a healthy subject.
  • these immunocompromised subjects may develop bloodstream infections caused by aerobic bacilli, anaerobes, and fungi.
  • Predisposing factors for septic shock include diabetes mellirus; cirrhosis; leukopenic states, especially those associated with underlying neoplasms or treatment with cytotoxic agents; antecedent infection in the urinary, biliary, or GI tracts; invasive devices, including catheters, drainage tubes, and other foreign materials; and prior treatment with antibiotics, corticosteroids, or ventilatory devices.
  • Septic shock occurs more often in newborns, subjects > 35 yr, pregnant women, and those seriously immunocompromised by underlying diseases or iatrogenic complications of treatment.
  • the above-described method is used for the prophylaxis of bacteremia, sepsis, septic shock, related conditions or their consequences.
  • the disorder is characterized by a risk of endotoxemia resulting from the use of antibiotic and the subsequent release of endotoxin, as well as positively identified bacteremia.
  • a person at risk for developing bacteremia, sepsis, septic shock and/or related conditions is a person that is considered to be immunocompromised and/or chronically ill.
  • the immunocompromised subject who is, at the time of bacterial exposure, has a preexisting condition that reduces one or more mechanisms for normal defense against infection.
  • the immunocompromised condition may be due to a defect or dysfunction of the immune system or to other factors that heighten susceptibility to infection, for example immunosuppressive agents.
  • the lactoferrin composition can reduce any of the following: the levels of circulating bacteria, the risk of the subject developing sepsis, septic shock, organ failure, and decrease the morbidity and mortality associated with bacteremia.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the risk of developing bacteremia and minimizing the effects of already existing bacteremia, sepsis, septic shock or related conditions.
  • the amount of the lactoferrin in the composition may vary from about 1 mg to about 100 g.
  • the guiding principle in the use of the lactoferrin composition is to administer the treatment at the earliest signs of bacteremia, sepsis or septic shock being developed to attenuate the development of bacteremia and to reduce the extent of organ damage that results from sepsis and septic shock.
  • the improvement is any observable or measurable improvement.
  • a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate levels of bacteria in circulation.
  • an improvement can consist of any of the following, for example, decrease in the levels of circulating bacteria, attenuating the development of sepsis, attenuating the development of septic shock, attenuating the development of organ failure, decreasing morbidity associated with bacteremia and decreasing mortality (death) associated with bacteremia.
  • the lactoferrin composition after administration of the lactoferrin composition, if any of the above conditions improve, then the amount of the lactoferrin composition is considered to be an effective amount. Yet further, administration of the lactoferrin composition will also attenuate the development of sepsis, septic shock and other conditions related thereto.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the severity of sepsis or septic shock.
  • an improvement can consist of any of the following, for example, decreasing mortality, decreasing morbidity, attenuating the development organ failure, decreasing days of hospitalization, decreasing or eliminating days of intensive care such as in an intensive care unit, decreasing or eliminating the use of supportive care such as a mechanical ventilator or decreasing the incidence of sequelae such as ARDS. Survival in patients with organ failure at baseline and prevention and reversal of organ failure are also evaluated.
  • the lactoferrin composition if any of the above conditions improve, then the amount the lactoferrin composition are considered to be an effective amount.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the severity of ALI or ARDS.
  • an improvement can consist of any of the following, for example, decrease in mortality, attenuating the development organ failure, decreasing days of hospitalization, decreasing or eliminating days of intensive care such as in an intensive care unit, or decreasing or eliminating the use of supportive care such as a mechanical ventilator or PaO 2 ZFiO 2 ratios.
  • compositions and methods of the invention may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment or prevention of bacteremia, sepsis, septic shock and related conditions, for example known agents to treat bacterial infections, e.g., antibiotics, known agents for the treatment of sepsis, e.g., Drotrecogin alfa (activated) and agents to treat inflammation.
  • a conventional therapy or agent including but not limited to, a pharmacological therapeutic agent may be combined with the composition of the present invention.
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention can be administered in any order or combination.
  • one or more agents may be administered substantially simultaneously, or within about minutes to hours to days to weeks and any range derivable therein, prior to and/or after administering the composition.
  • compositions to a cell, tissue or organism may follow general protocols for the administration of cardiovascular therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
  • Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include an antimicrobial agent, an anti-sepsis agent, an antiinflammatory agent, an antithrombotic/fibrinolytic agent, a blood coagulant, an antiarrhythmic agent, an antihypertensive agent, a vasopressor, or agents to treat metabolic acidosis.
  • antimicrobial agents e.g., antibiotics are used in combination with the composition of the present invention.
  • an "antimicrobial agent” can be used in combination with the lactoferrin composition.
  • An antimicrobial agent may comprise an antibiotic, anti-fungal, and anti-viral agent.
  • Antibiotics inhibits the growth of microorganisms without damage to the host.
  • the antibiotic may inhibit cell wall synthesis, protein synthesis, nucleic acid synthesis, or alter cell membrane function.
  • Classes of antibiotics that can possibly be used in conjunction with the peptide include, but are not limited to, macrolides (i.e., erythromycin), penicillins (i.e., nafcillin), cephalosporins (i.e., cefazolin), carbepenems (i.e., imipenem, aztreonam), other beta-lactam antibiotics, beta-lactam inhibitors (i.e., sulbactam), oxalines (i.e.
  • linezolid aminoglycosides (i.e., gentamicin), chloramphenicol, sufonamides (i.e., sulfamethoxazole), glycopeptides (i.e., vancomycin), quinolones (i.e., ciprofloxacin), tetracyclines (i.e., minocycline), fusidic acid, trimethoprim, metronidazole, clindamycin, mupirocin, rifamycins (i.e., rifampin), streptogramins (i.e., quinupristin and dalfopristin) lipoprotein (i.e., daptomycin), polyenes (i.e., amphotericin B), azoles (i.e., fluconazole), and echinocandins (i.e., caspofungin acetate).
  • aminoglycosides i.
  • antibiotics examples include, but are not limited to, erythromycin, nafcillin, cefazolin, imipenem, aztreonam, gentamicin, sulfamethoxazole, vancomycin, ciprofloxacin, trimethoprim, rifampin, metronidazole, clindamycin, teicoplanin, mupirocin, azithromycin, clarithromycin, ofloxacin, lomefloxacin, norfloxacin, nalidixic acid, sparfloxacin, pefloxacin, amifloxacin, gatifloxacin, moxifloxacin, gemifloxacin, enoxacin, fleroxacin, minocycline, linezolid, temafloxacin, tosufloxacin, clinafloxacin, sulbactam, clavulanic acid, amphotericin B, flucon
  • Anti-viral agents can also be used in combination the lactoferrin composition to treat and/or prevent a viral infection or disease.
  • Such anti-viral agents include, but are not limited to protease inhibitors (e.g., saquinavir, ritonavir, amprenavir), reverse transcriptase inhibitors (e.g., azidothymidine (AZT), lamioridine (3TC), dideoxyinosine (ddl)), dideoxycytidine (ddC), zidovudine), nucleoside analogs (e.g., acyclovir, penciclovir).
  • protease inhibitors e.g., saquinavir, ritonavir, amprenavir
  • reverse transcriptase inhibitors e.g., azidothymidine (AZT), lamioridine (3TC), dideoxyinosine (ddl)
  • ddC dideoxycyt
  • anti-fungal agents can be used in combination with the peptide compositon to treat and/or prevent a fungal infection.
  • anti-fungal agents include, amphotericin B (Amphocin®, Fungizone®), butoconazole (Femstat®), clotrimazole (Mycelex®, Gyne-Lotrimin®, Lotrimin®, Lotrisone®), fluconazole (Diflucan®), flucytosine (Ancobon®), griseofulvin (Fulvicin P/G®, Grifulvin V®, Gris-PEG®), itraconazole (Sporanox®), ketoconazole (Nizoral®), miconazole (Femizol-M®, Monistat®), nystatin (Mycostatin®), terbinafine (Lamisil®), terconazole (Terazol®), or tioconazole (Vagistat®).
  • Anti-sepsis agents include, but are not limited to Drotrecogin alfa (activated). Agents used for the treatment of ALI and ARDS include but are not limited to intra- pulmonary instillation of surfactants, and leukotriene modifiers. Anti-inflammatory agents include, but are not limited to non-sterodial anti-inflammatory agents (e.g., naproxen, ibuprofen, celeoxib) and sterodial anti-inflammatory agents (e.g., glucocorticoids).
  • non-sterodial anti-inflammatory agents e.g., naproxen, ibuprofen, celeoxib
  • sterodial anti-inflammatory agents e.g., glucocorticoids
  • Non-limiting examples of non-pharmacologic interventions that may be used in the present invention include supportive care such as organ support in sepsis and septic shock and low tidal volume ventilation protocols in ALI and ARDS.
  • supportive care such as organ support in sepsis and septic shock and low tidal volume ventilation protocols in ALI and ARDS.
  • composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced pain which may be acute or chronic in nature.
  • causes of acute pain include, but are not limited to pain following trauma such as a blunt or sharp injury, bullet wounds, or surgery.
  • causes of chronic pain include, but are not limited to pain associated with chronic diseases such as cancer, arthritis, inflammatory disease, chronic wounds, cardiovascular accidents, disorders of the spinal chord or central nervous system, and recovery from surgery.
  • chronic diseases such as cancer, arthritis, inflammatory disease, chronic wounds, cardiovascular accidents, disorders of the spinal chord or central nervous system, and recovery from surgery.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate pain.
  • a subject is administered a therapeutically effective amount of a lactoferrin composition so that the subject has an improvement in the parameters relating to pain including subjective measures such as pain scores and grading and objective measures including use of additional pain medications.
  • the improvement is any observable or measurable improvement.
  • a treatment may improve the patient condition, but may not be a complete cure of the disease.
  • the amount of lactoferrin in the composition may vary from about 1 ng to about 100 g each day.
  • the lactoferrin related lactoferrin composition is administered to a subject to modulate chronic or acute pain in the subject. Modulation of pain is achieved by administering an effective amount of the composition to decrease, reduce, inhibit or abrogate pain.
  • lactoferrin stimulates or enhances cytokine production or activity.
  • lactoferrin can enhance the production or activity of TNF- ⁇ resulting in modulation of pain, acute or chronic, in the subject.
  • compositions and methods of the invention may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment of pain both directly and indirectly.
  • a conventional therapy or agent including but not limited to, a pharmacological therapeutic agent, a non-pharmacological pain management techniques or a combination thereof, may be combined with the composition of the present invention.
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks, hi embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention and agents are employed.
  • agents can be administered in any order or combination.
  • one or more agents may be administered within of from substantially simultaneously, about minutes to hours to days to weeks and any range derivable therein, prior to and/or after administering the composition.
  • compositions to a cell, tissue or organism may follow general protocols for the administration of pain therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
  • Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include non-steroidal anti-inflammatory drugs (NSAIDS) (e.g., aspirin, indomethacin, ibuprofen, naproxen, acetaminophen, and ketoprofen); opioid analgesics (e.g., codeine, morphine, methadone, demerol, fentanyl, oxymorphone, and levarphanol), second generation NSAIDs (e.g., COX-2 inhibitors); and anti-depressant drugs (e.g., tricyclin antidepressants, sedatives, tranquilizers, hypnotics, anti-histamines, and amphetamines).
  • NSAIDS non-steroidal anti-inflammatory drugs
  • opioid analgesics e.g., codeine, morphine, methadone, demerol, fentanyl, oxymorphone, and levarphanol
  • a therapeutic agent may comprise a non-pharmacological pain management techniques
  • Non-pharmacological pain management techniques are well known to those of skill in the art, and may comprise, but are not limited to acupuncture or acupressure, local anesthesia/analgesia, regional anesthesia/analgesia (e.g., epidural or spinal analgesia/anesthesia), general anesthesia/analgesia (e.g., intravenous anesthetics or opioid pump) and chiropractic.
  • Non-pharmacological pain-reduction agents include, but are not limited to devil's, capsaicin, menthold or L-phenylalanine.
  • the composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced or is at risk of developing cardiovascular disease.
  • Risk factors include, but are not limited to elevated levels of cholesterol or CRP.
  • One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce circulating levels of total cholesterol or triglycerides or cardiovascular inflammation.
  • Cardiovascular diseases and/or disorders include, but are not limited to, diseases and/or disorders of the pericardium, heart valves (e.g., incompetent valves, stenosed valves, Rheumatic heart disease, mitral valve prolapse, aortic regurgitation), myocardium (e.g., coronary artery disease, myocardial infarction, heart failure, ischemic heart disease, angina) blood vessels (e.g., hypertension, arteriosclerosis, aneurysm) or veins (e.g., varicose veins, hemorrhoids).
  • the cardiovascular disease is atherosclerosis.
  • the lactoferrin composition is administered to a subject suffering from or at risk for developing atherosclerosis.
  • the lactoferrin composition modulates or reduces the severity and/or incidence of atherosclerosis.
  • Prophylactic treatment can be administered to those subjects at risk for developing atherosclerosis.
  • One risk factor is an atherogenic lipoprotein profile. For example, a ratio of serum cholesterol to high density lipoproteins of above 5:1 indicates a higher than average risk of developing atherosclerosis.
  • Other factors indicating increased risk for atherosclerosis include a serum cholesterol level of above 240 mg/dl; a high density lipoprotein level below about 35 mg/dl; and a low density lipoprotein level above about 160 mg/dl.
  • Another embodiment includes treating a human subject with an elevated level of circulating total cholesterol or CRP according to the then medically established guidelines. It is contemplated that the composition of the present invention reduces or attenuates the levels of circulating total cholesterol, low density lipoproteins or very low density lipoproteins. It is contemplated that the composition of the present invention can interfere with how cholesterol enters the circulation either via absorption from food (exogenous pathway) or synthesis by the liver (endogenous pathway).
  • Another embodiment includes treating a human subject having dyslipedemia.
  • Dyslipidemias are disorders of lipoprotein metabolism, including lipoprotein overproduction or deficiency. These disorders may be manifested by elevation of the serum total cholesterol, low-density lipoprotein (LDL) cholesterol and triglyceride concentrations, and a decrease in the high-density lipoprotein (HDL) cholesterol concentration.
  • LDL low-density lipoprotein
  • HDL high-density lipoprotein
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate cardiovascular disease.
  • a subject is administered a therapeutically effective amount of a lactoferrin composition so that the subject has an improvement in the parameters relating to cardiovascular disease including circulating levels of total cholesterol, HDL, LDL, VLDL, trigylcerides and C- reactive protein (CRP).
  • the amount of lactoferrin in the composition may vary from about 1 ng to about 20 g.
  • the composition that is orally administered contains the range of 0.5 g to 5 g of lactoferrin per day.
  • the improvement is any observable or measurable improvement.
  • a treatment may improve the patient condition, but may not be a complete cure of the disease.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate excess amounts of cholesterol levels in circulation.
  • a subject requires treatment for cholesterol levels based upon any of the following situations: LDL of 160 mg/ml or greater; LDL of 130-159 mg/ml and also have two or more cardiovascular risk factors; LDL of 100 mg/ml or greater in subjects with coronary heart disease (CHD); triglycerides of 200 mg/dl or higher; total cholesterol of 240 mg/dl or higher or HDL of less than 40 mg/dl.
  • CHD coronary heart disease
  • triglycerides 200 mg/dl or higher
  • FIG. 1 Another embodiment is a method of reducing vascular inflammation by administering the composition of the present invention.
  • Vascular inflammation can be tied to a number of the underlying processes contributing to atherosclerosis which include endothelial dysfunction, vascular proliferation and matrix alteration. Recent studies have emphasized the involvement of inflammation in mediating all stages of atherosclerosis. Vascular inflammation is thought to be a consequence of damage to the vascular endothelium and may also involve the proliferation of vascular smooth muscle cells (vsmcs). One precursor of lesion development in humans may be focal accumulation of vsmcs within the intima.
  • vsmcs vascular smooth muscle cells
  • vsmcs may contribute to the development of the atheroma through the production of pro-inflammatory mediators such as monocyte chemoattractant protein 1 and vascular cell adhesion molecule, and through the synthesis of matrix molecules required for the retention of lipoproteins. Inflammation of the vascular endothelium and proliferation of vsmcs may also impact the stability of the plaque through the formation of a firm fibrous cap. Indeed, in lipid-laden lesions in which the fibrous cap is thin and weak, there is evidence of vsmc apoptosis, especially at the "shoulder" region, associated with inflammation.
  • the local inflammatory milieu can induce expression of collagenase and inhibit expression of proteolytic inhibitors, thus rendering the fibrous cap weak and susceptible to rupture.
  • Lactoferrin having known anti-inflammatory properties, may thus serve to inhibit the underlying processes associated with the development of atherosclerosis.
  • the composition may also reduce vascular spasms or vascular hyper-reactivity.
  • Vascular spasms are a sudden, brief tightening of a blood vessel, which can temporarily reduce blood flow to tissues supplied by that vessel.
  • the composition may also promote endothelial integrity or healing.
  • Endothelia are the layer of cells lining the blood vessels. Endothelial dysfunction most commonly refers to impairment of endothelium-dependent vasodilation and widespread abnormalities in endothelial integrity and homeostasis. It is believed that HDLs help maintain endothelial integrity, facilitate vascular relaxation, inhibit blood cell adhesion to vascular endothelium, reduce platelet aggregability and coagulation, and may favor fibrinolysis. The integrity or completeness of the endothelia lining of the vessels is important to preventing/treating the development of plaques and atherosclerosis. Thus, it is envisioned that the composition of the present invention will promote or modulate endothelial integrity or healing.
  • another embodiment is a method of preventing a cardiovascular disease in a subject at risk for developing a cardiovascular disease comprising the step of administering to the subject a lactoferrin composition in an amount sufficient to result in prophylaxis of the cardiovascular disease in the subject.
  • the cardiovascular disease is atherosclerosis. It is envisioned that the composition not only possess therapeutic benefits for those subjects suffering from cardiovascular diseases, but also possess prophylactic properties for those subjects at risk for developing cardiovascular disease. A subject at risk may or may not be cognizant of their disease state or potential disease state and may or may not be aware that they are need of treatment.
  • the composition can reduce any of the following: the levels of circulating total cholesterol, low density lipoproteins (LDL), very low density lipoproteins (VLDL), levels of vascular inflammation, circulating C-reactive protein (CRP), triglycerides, and the proliferation of vascular smooth muscle cells in the subject. Yet further, the composition may also increase the levels of circulating high density lipoproteins (HDL). [0301] In order to increase the effectiveness of the composition, it may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment or prevention of cardiovascular disease or disorder, for example known agents to treat or prevent atherosclerosis.
  • a known agent effective in the treatment or prevention of cardiovascular disease or disorder for example known agents to treat or prevent atherosclerosis.
  • a conventional therapy or agent including but not limited to, a pharmacological therapeutic agent, a surgical therapeutic agent (e.g., a surgical procedure) or a combination thereof, may be combined with the composition of the present invention.
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention and agents are employed.
  • agents can be administered in any order or combination.
  • one or more agents may be administered substantially simultaneously, ⁇ ivithin about minutes to hours to " " days to weeks and any range derivable therein, prior to and/or after administering the composition.
  • compositions to a cell, tissue or organism may follow general protocols for the administration of cardiovascular therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
  • Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include an antihyperlipoproteinemic agent, an antiarteriosclerotic agent, an anti-cholesterol agent, an anti-inflammatory agent, an antithrombotic/fibrinolytic agent, a blood coagulant, an antiarrhythmic agent, an antihypertensive agent, or a vasopressor.
  • anti-cholesterolemic agents are used in combination with the composition of the present invention.
  • Anti-cholesterol agents include but are not limited to HMG-CoA Reductase inhibitors, cholesterol absorption inhibitors, bile acid sequestrants, nicotinic acid and derivatives thereof, fibric acid and derivatives thereof.
  • HMG-CoA Reductase inhibitors include statins, for example, but not limited to atorvastatin calcium (Lipitor®), cerivastatin sodium (Baycol®), fluvastatin sodium (Lescol®), lovastatin (Advicor®), pravastatin sodium (Pravachol®), and simvastatin (Zocor®).
  • Agents known to reduce the absorption of ingested cholesterol include, for example, Zetia®.
  • Bile acid sequestrants include, but are not limited to cholestyramine, cholestipol and colesevalam.
  • Other anti-cholesterol agents include fibric acids and derivatives thereof (e.g., gemfibrozil, fenofibrate and clofibrate); nicotinic acids and derivatives thereof (e.g., antibiotic anaspasmodic acid, for example niaspan.
  • Anti-inflammatory agents include, but are not limited to non-sterodial anti-inflammatory agents (e.g., naproxen, ibuprofen, celeoxib) and sterodial anti-inflammatory agents (e.g., glucocorticoids).
  • a therapeutic agent may comprise a surgery of some type, which includes, for example, preventative, diagnostic or staging, curative and palliative surgery.
  • Surgery and in particular a curative surgery, may be used in conjunction with other therapies, such as the present invention and one or more other agents.
  • Such surgical therapeutic agents for cardiovascular diseases and disorders are well known to those of skill in the art, and may comprise, but are not limited to, performing surgery on an organism, providing a cardiovascular mechanical prostheses, angioplasty, coronary artery reperfusion, catheter ablation, providing an implantable cardioverter defibrillator to the subject, mechanical circulatory support or a combination thereof.
  • a mechanical circulatory support that may be used in the present invention comprise an intra-aortic balloon counterpulsation, left ventricular assist device or combination thereof.
  • a subject that is suspected of an autoimmune disease or a subject suffering from an autoimmune disease is treated with the lactoferrin composition of the present invention.
  • autoimmune disorders The pathogenesis of autoimmune disorders is mediated by the aberrant activation, differentiation and trafficking of leukocytes in response to tissue self-antigens. Autoimmune reactions can often have fatal consequences. They can cause the destruction of vital tissue which manifests in the development of chronic autoimmune diseases.
  • the driving forces in the progression of these diseases are autoreactive immune cells. In most cases, these autoreactive cells are CD4 + T-cells which somehow escaped the self-tolerance control mechanisms of the immune system. In non-pathogenic situations, CD4+ T-cells act as T helper cells, which control or mediate the activation and differentiation of other immune cells such as cytotoxic CD 8+ T- cells, NK cells, granulocytes, macrophages and B-cells.
  • CD4 + T- cells are no longer just mediators, but are rather key players of the autoimmune response. In particular, they are either directly or indirectly responsible for the characteristic tissue destruction that occurs in autoimmune diseases, which is triggered by the recognition of autoantigens.
  • autoantigens are derived from self-proteins of the attacked tissue and are presented to the autoreactive CD4+ T- cells by antigen-presenting cells (APCs), such as macrophages, dendritic cells or B-cells.
  • APCs antigen-presenting cells
  • B-cells antigen-presenting cells
  • aberrant T cell-dependent or T cell- independent antibody production by activated B cells plays a critical role in initiating and amplifying tissue damage in autoimmune disorders.
  • Such autoimmune disorders include, but are not limited to, the following: Alopecia Areata, Ankylosing Spondylitis, Antiphospholipid Syndrome, aplastic anemia, myelodysplastic syndromes, paroxysmal nocturnal hemoglobulinemia, pure red cell aplasia, chronic neutropenias, amegakaryocytic thrombocytopenia, antiphospholipid syndromes, autoimmune thrombocytopenia, autoimmune hemolytic syndromes, antiphospholipid syndromes, autoimmune gastritis, achlorhydria, Autoimmune Addison's Disease, Autoimmune Diabetes, Autoimmune Hemolytic Anemia, Autoimmune Hepatitis, Autoimmune hypophysitis, Autoimmune orchiditis, autoimmune ovarian failure, Behcet's Disease, Bullous Pemphigoid, Cardiomyopathy, Celiac Sprue-Dermatitis, Cicatrical pemphi
  • the autoimmune disease that is treated includes, but is not limited to The present invention is also designed to treat any autoimmune disease including but not limited to Active Chronic Hepatitis, Addison's Disease, Anti-phospholipid Syndrome, Atopic, Allergy, Autoimmune, Atrophic Gastritis, Achlorhydra Autoimmune, Celiac Disease, Crohn's Disease, Cushings Syndrome, Dermatomyositis, (Type I) Diabetes, Discoid Lupus Erythematosis, Goodpasture's Syndrome, Grave's Disease, Hashimoto's Thyroiditis, Idiopathic Adrenal Atrophy, Idiopathic Thrombocytopenia, Insulin-dependent Diabetes, Lambert-Eaton Syndrome, Lupoid Hepatitis, Lymphopenia (some cases), Mixed Connective Tissue Disease, Multiple Sclerosis, Pemphigoid, Pemphigus Vulgaris, Pernicious Anema, Phacogenic Uveitis, Polyart
  • the composition of the present invention is administered to a subject suffering from graft versus host disease (GVHD), organ transplant rejection, autoimmune hepatitis, primary biliary cirrhosis, autoimmune cholangitis, primary sclerosing cholangitis, irritable bowel syndrome (IBS), multiple sclerosis (MS), chronic granulomatous disease, ankylosing spondylitis, scleroderma, polymyositis, (dermato)myositis, systemic vasculitis, systemic lupus erythematosus (SLE), Chrohn's disease, insulin-dependent diabetes (type 1) or ulcerative colitis.
  • GVHD graft versus host disease
  • organ transplant rejection autoimmune hepatitis
  • primary biliary cirrhosis autoimmune cholangitis
  • primary sclerosing cholangitis primary sclerosing cholangitis
  • IBS multiple sclerosis
  • chronic granulomatous disease
  • the composition provided in any of the above-described pharmaceutical carriers is administered as a therapeutic drug to a subject _who has undergone organ transplant (kidney, heart, lung, liver, pancreas, bone marrow, ⁇ peripheral stem cells) and/or as a prophylactic drug to organ and/or tissue transplant, for example bone marrow (BM) or peripheral stem cell donor cells, to prevent the development of GVHD in recipients and/or to prevent or attenuate transplant rejection in the recipients.
  • organ transplant kidney, heart, lung, liver, pancreas, bone marrow, ⁇ peripheral stem cells
  • BM bone marrow
  • peripheral stem cell donor cells for example bone marrow
  • One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce chronic allograft rejection and toxicity associated with standard therapy, or the development of GVHD.
  • a further embodiment is treating, preventing or reducing the risk of developing graft-versus-host-disease by administering a lactoferrin composition to the donor organ or donor tissue prior to transplantation into the recipient. It is envisioned that administering the composition to the donor tissue/organ will attenuate the immune cells in the donor/organ and prevent the development of the immune response that is mounted against the recipient's tissue, thus preventing or attenuating GVHD.
  • the recipient and the donor organ/tissue can be treated with the composition of the present invention.
  • Additional embodiments of the present invention include a method of treating, preventing or attenuating the severity of tissue or organ transplant rejection in a recipient comprising the step of administering to the donor a lactoferrin composition in an amount sufficient to attenuate the tissue or organ transplant rejection in the recipient. It is envisioned that the composition reduces allogeneic immune responses in the recipient. The composition can also be administered to both the recipient and the donor.
  • Another embodiment is a method of treating, preventing or attenuating the severity of xenograft tissue or xenograft organ transplant rejection in a recipient comprising the step of administering to the xenograft donor a composition in an amount sufficient to attenuate the tissue or organ transplant rejection in the recipient.
  • the composition can also be administered to both the recipient and the xenograft donor.
  • progenitor cells or stem cells may be isolated from at least one organ, cell, tissue or organism.
  • Stem cells can be isolated from embryonic or nonembryonic donors.
  • the tissues from which the stem cells can be isolated include, for example, but are not limited to the bone marrow, the spleen, the liver, peripheral blood, umbilical cord tissue, umbilical cord blood, adipose tissue or skin.
  • tissue grafts may be used in the present invention. The tissue may be part or separated from an organism.
  • a tissue may comprise, but is not limited to, adipocytes, alveolar, ameloblasts, axon, basal cells, blood (e.g., lymphocytes), blood vessel, bone, bone marrow, peripheral stem cells, brain, breast, cartilage, cervix, colon, cornea, embryonic, endometrium, endothelial, epithelial, esophagus, facia, fibroblast, follicular, ganglion cells, glial cells, goblet cells, kidney, liver, lung, lymph node, muscle, neuron, ovaries, pancreas, peripheral blood, prostate, skin, skin, small intestine, spleen, stem cells, stomach, testes, or ascite tissue.
  • an organ is the graft.
  • Organs are comprised of tissues having a special function.
  • Exemplary organs that are used in grafts in the present invention include, but are not limited to heart, kidney, pancreas, lung, or liver.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate chronic allograft rejection and toxicity related to standard therapy.
  • the amount of lactoferrin in the composition may vary from about 1 ng to about 100 g, more preferably, lmg to about 2Og.
  • the composition that is orally administered contains the range of 0.1 g to 5 g of lactoferrin per day.
  • the improvement is any observable or measurable improvement.
  • a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate levels of an immune response against a graft in the recipient.
  • an improvement can consist of any of the following, for example, increased function of the graft, for example, increased urine output for kidney grafts or decreased jaundice for liver grafts; reduction in inflammation; reduction in general discomfort of the recipient; an overall increased tolerance for the graft.
  • the lactoferrin composition if any of the above conditions improve, then the amount is considered to be an effective amount.
  • GHVD is also any observable or measurable improvement.
  • a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease.
  • the composition is administered in an effective amount-to decrease, reduce, inhibit or abrogate levels of immune response from the donor's cells, tissue and/or organ against the host's tissues.
  • GVHD can be acute or chronic or mild or severe. Improvements in acute symptoms include any of the following, for example, decrease skin rash, decrease diarrhea, increase in liver function, a decrease in susceptibility to infection.
  • Improvements in chronic symptoms include, but are not limited to decrease skin rash, decrease dermatitis, decrease hair loss, a decrease liver damage, decrease dry eyes and mouth, a decrease susceptibility to infections and decrease lung and/or gastrointestinal disorders.
  • the amount is considered to be an effective amount.
  • a further embodiment is a method of regulating a mucosal immune response in the gastrointestinal tract or a systemic immune response in a subject comprising the step of administering a lactoferrin related lactoferrin composition to said subject the composition of the present invention.
  • composition stimulates MIP-3 ⁇ and interleukin-18 in the gastrointestinal tract, which regulates immune responses.
  • interleukin-18 modulates both Thl/Th2 responses.
  • IL- 18 plays an important role in allogeneic stem cell transplantation. Pre-treatment of allogeneic donors with IL- 18 significantly improves survival and reduces clinical and pathological indices of acute GVHD in BMT recipients.
  • cytokines may also be enhanced or repressed for example, but not limited to IL-I 5 IL-2, IL-6, ILlO, IL- 12 and GM-CSF. It is also envisioned that lactoferrin stimulates Thl/Th2 type-responses through the induction and/or repression of ThI and Th2 cytokines.
  • compositions of the present invention In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine these compositions with standard therapy.
  • known immunosuppressant agents are used in combination with the composition of the present invention.
  • agents known to prevent organ rejection are T cell modifiers such as cyclosporine (Neoral®, Sandimmune®), prednisone (Novo Prednisone®, Apo Prednisone®), azathioprine (Imuran®), tacrolimus or FK506 (Prograf®), mycophenolate mofetil (CellCept®), OKT3 (Muromorab CO3®, Orthoclone®), ATGAM® & Thymoglobulin® or serine-threonine phosphatase calcineurin (CN) inhibitors.
  • the standard or approved treatment of GVHD which is high doses corticosteroids, primarily high-dose methylprednisolone, is used in combination with the lacto
  • therapies for preconditioning and prophylaxis of GVHD include, but are not limited to total body irradiation, cytosine arabinoside, L-phenylalanine mustard, cyclophosphamide, etoposide, triethylene thiophosphoramide, antithymocyte globulin, bisulfan, tacrolimus, methylprednisolone, cyclosporin, or methotrexate.
  • cytokine inhibitors/antagonists e.g., anti- TNF ⁇ antibody
  • IL-I receptor antagonist e.g., IL-I receptor antagonist
  • recombinant IL-I receptor e.g., inhibitors of T-cell activation
  • antimetabolites e.g., mycophenolate mofetil
  • anti-CD3 antibody muromonab, OKT3
  • anti-CD25 antibody e.g., OKT3
  • anti-IL2 receptor monoclonal antibody daclizumab e.g., extracorporeal photopheresis using ex vivo 8-methoxypsoralen
  • anti-thymocyte globulins Thymoglobulin® or ATGAM®
  • ABX-CBL or CBL-I ABX-CBL or CBL-I
  • Visilizumab Nuvion®
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention are employed.
  • agents can be administered in any order or combination.
  • the composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced or is at high risk of having diabetes mellitus.
  • the composition of the present invention may be administered to a subject to regulate diabetes mellitus.
  • the composition modulates at least one symptom of diabetes mellitus, for example, decrease blood glucose or modulate blood insulin levels.
  • Risk factors for type I diabetes include islet-cell antibodies and those of type 2 or gestational diabetes include inactivity, obesity, siblings with diabetes, and history of diabetes during pregnancy.
  • One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce circulating levels of glucose.
  • the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate high glucose, or to reduce total body weight, glycosylated hemoglobin (HbAIc), or blood pressure or to modulate blood insulin levels.
  • HbAIc glycosylated hemoglobin
  • successful reduction of hyperglycemia by the lactoferrin composition may be manifested by the fasting plasma glucose level falling below 126 mg/dL, the 2-hour plasma glucose level during an oral glucose tolerance test (OGTT ) falling below below 200 mg/dL, or if a random plasma glucose determination reading below 200 mg/dL in a symptomatic individual.
  • a successful reduction of hyperglycemia by the lactoferrin composition may be manifested by the fasting plasma glucose falling below 110 mg/dL and/or the 2-hour plasma glucose on the OGTT falling below between 140 mg/dL.
  • the amount of lactoferrin in the composition may vary from about 0.1 ⁇ g to about 100 g, more preferably, from about 1 ⁇ g to about 20 g, or any range therebetween.
  • the composition that is administered contains the range of 0.1 g to 5 g of lactoferrin per day.
  • Glycohemoglobin (or glycosylated hemoglobin) is measured to monitor or accurately record blood glucose levels, and it is not influenced by acute changes in blood glucose or by the interval since the last meal. Glycohemoglobin is formed when glucose reacts non- enzymatically with the hemoglobin A molecule and is composed of several fractions, the major one being Hb AIc . Total glycohemoglobin (Hb Al) and Hb AIc (expressed as the percentage of total hemoglobin) vary in proportion to the average level of glucose over the lifespan of the red blood cell (RBC), thereby providing an index of glycemic control.
  • RBC red blood cell
  • Further aspects of the invention include reducing blood glucose in a patient suffering from diabetes mellitus by administering to a subject an effective amount of the composition such that the amount of the lactoferrin composition modulates blood glucose.
  • the blood glucose is monitored by the level of glycosylated hemoglobin (HbAIc).
  • Another aspect is a method of modulating blood insulin in a patient suffering from diabetes mellitus by administering to a subject an effective amount of the composition such that the amount modulates blood insulin.
  • Modulating blood insulin includes reducing or maintaining blood insulin levels or increasing blood insulin levels.
  • Modulating blood insulin levels in the present invention includes increasing blood insulin levels in insulin- dependent diabetes (Type I) or decreasing or maintaining insulin levels in non-insulin-dependent diabetes (Type II).
  • compositions In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine the compositions with an additional agent.
  • known diabetes agents are used in combination with the composition of the present invention.
  • Other antidiabetic agents may also include, a weight loss regimen and/or a diet alteration.
  • composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention are employed.
  • agents can be administered in any order or combination.
  • the petpide composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a fragile bone condition.
  • the present invention is designed for the treatment of any type of fragile bone condition or any disorder relating to bone loss including but not limited to osteoporosis, age-associated osteoporosis, postmenopausal osteoporosis, osteitis deformans (Paget' s disease), osteogenesis imperfecta (brittle bones), and osteopetrosis.
  • any type of fragile bone condition or any disorder relating to bone loss including but not limited to osteoporosis, age-associated osteoporosis, postmenopausal osteoporosis, osteitis deformans (Paget' s disease), osteogenesis imperfecta (brittle bones), and osteopetrosis.
  • Bone loss disorders arise from an imbalance in the formation of new healthy bone and the resorption of old bone, skewed toward a net loss of bone tissue.
  • This bone loss includes a decrease in both mineral content and protein matrix components of the bone, and leads to an increased fracture rate of, predominantly, femoral bones and bones in the forearm and vertebrae.
  • fractures in turn, lead to an increase in general morbidity, a marked loss of stature and mobility, and, in many cases, an increase in mortality resulting from complications.
  • Bone loss occurs in a wide range of subjects, including postmenopausal women, patients who have undergone hysterectomy, patients who are undergoing or have undergone long-term administration of corticosteroids, patients suffering from Cushing's syndrome, and patients having gonadal dysgensis.
  • osteoporosis a major debilitating disease whose prominent feature is the loss of bone mass (decreased density and enlargement of bone spaces) without a reduction in bone volume, producing porosity and fragility.
  • Osteoporosis is common in the elderly of both sexes but is more pronounced in postmenopausal women. Osteoporosis may occur as a primary disorder or as a secondary complication of several diseases. It is proposed that genetic factors determine the size of the bone mass achieved in young adulthood. With aging, the increased osteoclastic function and the slowing of osteoblastic activity induced by endocrine influences, particularly decreased estrogen levels, result in a net negative balance in the continued turnover of bone. Osteoporosis causes bone pain owing to microfractures; results in loss in height and stability of the vertebral column; and predisposes to fractures of femoral necks, wrists, and vertebrae. The condition remains asymptomatic until skeletal fragility is well advanced.
  • Paget' s disease is currently considered to be a slow paramyxoviral infection of osteoblasts and then osteoclasts. The condition is divided into an initial osteolytic stage, followed by a mixed osteolytic-osteoblastic stage, evolving ultimately into burn-out quiescent osteosclerotic stage. Because new bone formation in active disease is disordered and poorly mineralized, it is soft and porous, lacks structural stability, and is vulnerable to fracture or deformation under stress. Patients may demonstrate fractures, nerve compression, osteoarthritis, and skeletal deformities.
  • Osteogenesis imperfecta or brittle bones refers to a group of closely related genetic disorders caused by qualitative or quantitative abnormal synthesis of type I collagen, constituting about 90% of the matrix of bone. Syndromes range from one variant (type II) that is uniformly fatal in the perinatal period (from multiple bone fractures) to other variants marked by increased predisposition to fracture but compatible with survival. Morphologically the basic change in all is osteopenia or too little bone, with marked thinning of the cortices and rarefication of the trabeculae.
  • Osteopetrosis refers to a group of rare hereditary diseases characterized by overgrowth and sclerosis of boone, with marked thickening of the cortex and narrowing or filling of the medullary cavity impairing hematopoisesis. Despite too much bone, it is brittle and fractures easily. The autosomal recessive form is evident from birth, with anemia, neutropenia, infections and eventual death. The autosomal dominant form is benign but predisposes to fractures. Common to all forms is a hereditary defect in osteoclast function resulting in reduced bone resorption and enhanced net bone overgrowth.
  • lactoferrin composition is administered in an effective amount to stabilize or reduce bone fragility, to strengthen the bone, inducing the growth of healthy bone, modulate calcium levels, and modulate mineral accumulation in the skeleton.
  • the amount may vary from about 1 ⁇ g to about 100 g of lactoferrin composition.
  • the amount that is administered is in the range of 10 mg to 25 g.
  • the amount that is administered is in the range of 10 ⁇ g to 5g.
  • a dosage form a may contain a pharmaceutically acceptable non-toxic salt of the compounds which has a low degree of solubility in body fluids, for example, (a) an acid addition salt with a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene mono- or di-sulfonic acids, polygalacturonic acid, and the like; (b) a salt with a polyvalent metal cation such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed from e.g., N,N'-dibenzyl
  • the compounds of the present invention or, preferably, a relatively insoluble salt such as those just described may be formulated in a gel, for example, an aluminum monostearate gel with, e.g. sesame oil, suitable for injection.
  • Particularly preferred salts are zinc salts, zinc tannate salts, pamoate salts, and the like.
  • Another type of slow release depot formulation for injection would contain the compound or salt dispersed for encapsulation in a slow degrading, non-toxic, non-antigenic polymer such as a polylactic acid/polyglycolic acid polymer for example as described in U.S. Pat. No. 3,773,919.
  • the compounds or, preferably, relatively insoluble salts such as those described above may also be formulated in cholesterol matrix silastic pellets, particularly for use in animals. Additional slow release, depot or implant formulations, e.g. liposomes, are well known in the literature. See, for example, "Sustained and Controlled Release Drug Delivery Systems", J. R. Robinson ed., Marcel Dekker, Inc., N. Y., 1978.
  • a further embodiment is a method of promoting the stimulation or inhibition of cytokines, growth factors or mediators of bone growth or resorption.
  • growth factors include, but not limited to insulin-like growth factor- 1 (IGF-I), transforming growth factor-beta (TGF -b), and growth hormone releasing factor (GRF).
  • IGF-I insulin-like growth factor- 1
  • TGF -b transforming growth factor-beta
  • GRF growth hormone releasing factor
  • Mediators of bone growth or resorption include, but are not limited to parathyroid hormone (PTH), calcitonin or 1,25- dihydroxycholecalciferol (1 ,25(OH)2D3).
  • PTH parathyroid hormone
  • calcitonin 1,25- dihydroxycholecalciferol (1 ,25(OH)2D3
  • a further embodiment is a method of activating or inhibiting cells involved in bone remodeling including but not limited to osteoblasts, osteoclasts, chondrocytes, endothelial cells, and fibroblasts.
  • composition of the present invention may be desirable to combine the composition of the present invention with other -agents effective in the treatment of fragile bone diseases including but not limited to calcium and cholecalciferol (vitamin D) supplements, estrogen replacement therapy (ERT), raloxifene, calcitonin, bisphosphonates such as alendronate and cyclical etidronate, statins, recombinant human parathyroid hormone (rhPTH), insulin-like growth factor- 1 (IGF-I), and transforming growth factor-beta (TGF-I).
  • This process may involve administering the lactoferrin composition of the present invention and the agent(s) or multiple factor(s) at the same time.
  • compositions or pharmacological formulations that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the human lactoferrin composition and the other includes the second agent(s).
  • the composition of the present invention may precede or follow the other bone healing agent treatment by intervals ranging from minutes to weeks.
  • the other bone healing agent and the inventive composition are administered or applied separately, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and the composition would still be able to exert an advantageously combined effect on the condition.
  • composition of the present invention is administered to an immunosuppressed individual or an individual whose immune system is expected to be suppressed.
  • the subject method is suited for preventing infection and/or inflammation in immunosuppressed individuals or individuals whose immune systems are expected to be repressed.
  • the immunosuppressed subject exhibits neutropenia, or suffers from AIDS or any form of cancer including lymphoma and myeloma.
  • the subject undergoes or is expected to undergo a therapy selected from the group consisting of chemotherapy, radiotherapy, and autologous peripheral stem cell transplant.
  • An effective amount of the lactoferrin composition depends on the severity and/or course of immunosuppression, the patient's clinical history and response, and the discretion of the attending physician.
  • the composition is suitably administered to the patient at_ one time or over a series of treatments.
  • the initial candidate dosage may be administered to a patient.
  • the proper dosage and treatment regimen can be established by monitoring the progress of therapy using conventional techniques known to the people skilled of the art.
  • the amount of active ingredients that may be combined with the carrier materials to produce a single dosage form will vary depending upon the subject treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors, including the activity of the specific composition employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy, and can be determined by those skilled in the art.
  • the lactoferrin composition is administered prior to the inception neutropenia, more specifically Grade 3 or higher neutropenia and continued on the same schedule daily for the entire period during which the patient is at a high risk of neutropenic fever, infection or mucositis.
  • cytokines include interleukin-18 and GM- CSF in the gastrointestinal tract, which are known to enhance immune cells or stimulate production of immune cells.
  • Certain treatment regimens include administering the composition in which the concentration of lactoferrin that is provided is in the range of 1 g to about 100 g/ ml and the composition is a solution that is swished in the mouths of the subjects for a few minutes (e.g., about 4 minutes) and then swallowed.
  • This process can be repeated mutiple times a day, for example twice or more each day, for a total of three doses per 24 hour period.
  • the doses are taken immediately after meals (one each after breakfast, lunch and dinner), or at similarly spaced intervals throughout the day. Subjects are asked not to eat or drink anything for a least one hour after swallowing the dose.
  • the treatment may be terminated in the event of significant drug related adverse effects observed during one of the interim safety analyses.
  • the multiple administration of the lactoferrin composition each day can maximize the exposure of the oral cavity and gut to the drug.
  • patients an effective amount of the composition prophylactically for 21-28 days starting from the commencement of chemotherapy.
  • Daily dosing continues through the neutropenic phase until the earliest of the following: a) Absolute Neutrophil Count (ANC) is > 600 cells/ ⁇ L for three consecutive days, and mucositis (if present) has improved to Grade 2 or better for three consecutive days; or b) patient is discharged from the hospital or outpatient transplant setting.
  • a last-day-of-therapy evaluation is performed within 24 hours after the last dose.
  • the patients are also required to return for an end-of-treatment evaluation, to be performed 3-7 days after the last dose.
  • the treatment regimen in the present invention can prevent or attenuate infections in immunosuppressed patients, for example, patients undergoing aggressive chemotherapy or radiotherapy.
  • the treatment reduces incidence, duration, and severity, and prolongs the infection-free interval of neutropenic infection (neutropenic fever).
  • neutropenic fever is considered as an oral temperature of at least 38.0°C on at least two
  • Neutropenic fever is defined as fever that begins when the patient is neutropenic (ANC ⁇ 500).
  • the end of neutropenic fever is defined as the point at which the patient begins a period of 48 hours without fever (as defined above), whether or not the patient is still neutropenic.
  • Incidence of neutropenic fever is calculated as the number of patients experiencing one or more episodes of neutropenic fever. Onset of neutropenic fever is determined in two ways: (1) days between the transfusion and the first episode of neutropenic fever and (2) days between a neutrophil count ⁇ 1000/uL and the first episode of neutropenic fever.
  • Duration of neutropenic fever is defined as the number of days between the onset and the end of neutropenic fever. Severity of neutropenic fever is determined by the average temperature of the patient during the period of neutropenic fever. Infection-free interval is the period between the end of an infective fever and the onset of the next infective fever.
  • the present treatment also reduces the incidence, severity, and duration of oral and GI mucositis by improving all clinical criteria used for mucositis evaluation (pain, salivation, appetite, and oral inflammation). The incidence, duration, and severity of oral mucositis are assessed for each patient. Oral mucositis is graded using the Oral Mucositis Assessment Scale (OMAS). __ _ . - -
  • Prophylactic and concomitant use of antibiotics may be used during the treatment according to patient's conditions and usual standard of care in the medical profession.
  • Use of hematopoietic growth factors is not recommended routinely, but G-CSF may be used as part of the standard of clinical carte at the discretion of the doctors. All medications necessary for the patient's well being may also be administered.
  • the responses of the patients in the course of treatment are closely monitored by evaluating various response parameters.
  • the treatment protocol may be adjusted accordingly depending on the improvement of the patient's physical conditions.
  • the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a gastrointestinal disorder.
  • Gastrointestinal disorders include, but are not limited to, heartburn, bloating, postoperative ileus, abdominal pain and discomfort, early satiety, epigastric pain, nausea, vomiting, burbulence, regurgitation, intestinal pseudoobstruction, anal incontinence, gastroesophageal reflux disease, irritable bowel syndrome, ulcerative colitis, Crohn's disease, menstrual cramps, pancreatitis, spastic and interstitial cystitis and ulcers and the visceral pain associated therewith.
  • the lactoferrin composition is administered to a subject suffering from Crohn's disease, colitis, necrotizing entercolitis, endometriosis, irritable bowel syndrome, pancreatitis, periodontal disease, and ulcerative colitis.
  • the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a neurological disorder.
  • composition of the present invention when " administered to a subject suffering from a neurological disorder, stimulates the immune system thereby alleviating the neurological conditions.
  • the composition of the present invention can be useful with regard to the prevention, treatment, or amelioration of neurological, psychiatric, psychological, conscious state, behavioral, mood, and thought activity (unless otherwise indicated these will be collectively referred to herein as "neurological activity” which includes “psychological activity” or “psychiatric activity”).
  • neurological activity which includes “psychological activity” or “psychiatric activity”
  • a pathological or undesirable condition associated with the activity reference may be made to a neurological disorder which includes “psychiatric disorder” or “psychological disorder” instead of neurological activity or psychiatric or psychological activity.
  • a disorder such as a attention or cognitive disorders (e.g., Autistic Spectrum Disorders); mood disorder (e.g., major depressive disorder, bipolar disorder, and dysthymic disorder) or an anxiety disorder (e.g., panic disorder, posttraumatic stress disorder, obsessive-compulsive disorder and phobic disorder); neurodegenerative diseases (e.g., multiple sclerosis, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, sleep disorders, and chronic idiopathic demyelinating disease (CID)), one skilled in the art appreciates that the invention may also find application in conjunction with enhancing or diminishing any neurological or psychiatric function, not just an abnormality or disorder.
  • attention or cognitive disorders e.g., Autistic Spectrum Disorders
  • mood disorder e.g., major depressive disorder, bipolar disorder, and dysthymic disorder
  • an anxiety disorder
  • the lactoferrin related lactoferrin composition is administered to a subject suffering from multiple sclerosis, Alzheimer's disease, Parkinson's disease, muscular dystrophy, sleep or depression.
  • Neurological are diagnosed based upon the accumulation of physical, chemical, and historical behavioral data on each patient.
  • One of skill in the art is able to perform the appropriate examinations to accumulate such data.
  • One type of examination can include neurological examinations, which can include mental status evaluations, which can further include a psychiatric assessment.
  • Other types of examinations can include, but are not limited to, motor examination, cranial nerve examination, and neuropsychological tests (e.g., Minnesota Multiphasic Personality Inventory, Beck Depression Inventory, or Hamilton Rating Scale for Depression).
  • any improvement is based upon an improvement in any of the examinations that are used to diagnose a neurological disorder.
  • imaging techniques can be used to determine normal and abnormal brain function that can result in disorders.
  • Functional brain imaging allows for localization of specific normal and abnormal functioning of the nervous system. This includes electrical methods such as electroencephalography (EEG), magnetoencephalography (MEG), single photon emission computed tomography (SPECT), (SPECT), as well as metabolic and blood flow studies such as functional magnetic resonance imaging (fMRI), and positron emission tomography (PET) which can be utilized to localize brain function and dysfunction.
  • EEG electroencephalography
  • MEG magnetoencephalography
  • SPECT single photon emission computed tomography
  • SPECT single photon emission computed tomography
  • metabolic and blood flow studies such as functional magnetic resonance imaging (fMRI), and positron emission tomography (PET) which can be utilized to localize brain function and dysfunction.
  • fMRI functional magnetic resonance imaging
  • PET positron emission tomography
  • stimulating drugs comprise medications, anesthetic agents, synthetic or natural peptides or hormones, neurotransmitters, cytokines and other intracellular and intercellular chemical signals and messengers, and the like.
  • certain neurotransmitters, hormones, and other drugs are excitatory for some tissues, yet are inhibitory to other tissues.
  • a drug is referred to as an "excitatory” drug, this means that the drug is acting in an excitatory manner, although it may act in an inhibitory manner in other circumstances and/or locations.
  • an “inhibitory” drug is mentioned, this drug is acting in an inhibitory manner, although in other circumstances and/or locations, it may be an “excitatory” drug.
  • excitatory neurotransmitter agonists e.g., norepinephrine, epinephrine, glutamate, acetylcholine, serotonin, dopamine
  • agonists thereof and agents that act to increase levels of an excitatory neurotransmitter(s)
  • an excitatory neurotransmitter(s) e.g., edrophonium; Mestinon; trazodone; SSRIs (e.g., flouxetine, paroxetine, sertraline, citalopram and fluvoxamine); tricyclic antidepressants (e.g., imipramine, amitriptyline, doxepin, desipramine, trimipramine and nortriptyline), monoamine oxidase inhibitors (e.g., phenelzine, tranylcypromine, isocarboxasid)), generally have an excitatory effect on neural tissue, while inhibitory neurotransmitters
  • antagonists of inhibitory neurotransmitters e.g., bicuculline
  • agents that act to decrease levels of an inhibitory neurotransmitter(s) have been demonstrated to excite neural tissue, leading to increased neural activity.
  • excitatory neurotransmitter antagonists e.g., prazosin, and metoprolol
  • agents that decrease levels of excitatory neurotransmitters may inhibit neural activity.
  • lithium salts and anesthetics e.g., lidocane
  • antidepressants are administered in combination with the present invention to treat an affective disorder, such as depression.
  • antidepressants include, but are not limited to monoamine oxidase inhibitors, 5-HT1A agonists, selective 5-HT uptake inhibitors, imipramine or clomipramine.
  • the composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks.
  • composition of the present invention and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
  • compositions of the present invention are employed.
  • agents can be administered in any order or combination.
  • the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a hematological disorder.
  • Such disorders can include, but are not limited to anaemia, sickel cell anemia or cachexia.
  • the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having an ocular disorder.
  • ocular disorder include, but are not limited to conjunctivitis, dry eye disease, glaucoma, allergic eye disease, uveitis or ocular infection.
  • Dry eye disease or keratoconjunctivitis sicca affects about 4 million people in the U.S. of which about 1.5 million have moderate to severe disease. Globally, 60 million people use artificial tears (Damato et al. 1984, Hikichi et al. 1995, Bjerrum et al. 1997, Schein et al. 1997). Despite the high levels of incidence, there is currently no highly effective treatment. Development of therapeutic treatments has been hampered by the lack of knowledge regarding the etiology of the condition to its complexity. However, it has now been suggested that inflammation affecting the lacrimal gland and ocular surface underlies the pathophysiological process (Pflugfelder et al. 1986, Raphael et al.
  • lacxoierrin composition ot the present invention can be used to decrease the inflammatory response thereby treating the ocular disease or disorder.
  • the lactoferrin compositions are preferably formulated as topical ophthalmic suspensions or solutions, with a pH of about 4 to 8.
  • the establishment of a specific dosage regimen for each individual is left to the discretion of the clinicians.
  • the amount of lactoferrin will normally be contained in these formulations in an amount 0.01% to 5% by weight, but preferably in an amount of 0.05% to 2% and most preferably in an amount 0. 1 to 1.0% by weight.
  • the dosage form may be a solution, suspension microemulsion.
  • 1 to 2 drops of these formulations would be delivered to the surface of the eye 1 to 4 times per day according to the discretion of a skilled clinician.
  • the present invention is also designed to treat any fibrotic condition including but not limited to diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis (cirrhosis), and atherosclerosis.
  • any fibrotic condition including but not limited to diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis (cirrhosis), and atherosclerosis.
  • the present invention is also designed to treat any inflammatory condition, being non-infectious.
  • non-infectuious inflammatory conditions include but are not limited to rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ specific inflammatory diseases, glomerular disease, Sjogrens disease, or Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
  • a chemotactic modulator The migration of cells towards a source of a chemotactic modulator is tested in a chemotaxis or chemoattraction cell migration assay, including lactoferrin, chemokines, chemokine receptors, inflammatory or immune mediators, viral anti-chemokines, a mixture of casein and autologous serum, chemotactic C 5 fragment, and formyl-1-methionyl-l-leucine-l- phenylalanine (f-Met-Leu-Phe), bacterial factor derived from Escherichia coli, the simple peptide formylmethionylalanine, and activated human complement, carbamylcholine, phenylephrine, and cyclic guanosine 5 '-monophosphate, nucleosides and nucleotides, and prostaglandins.
  • Chemotactic modulators are tested alone or in combination with other chemotactic modulators to assess additive,
  • the different cell types used are deriyeji from healthy or diseased tissue and are from human, mouse or other species, including T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
  • T cell lines such as Jurkat
  • APCs antigen presenting cells
  • PBMCs peripheral blood mononuclear cells
  • splenocytes lymph node cells
  • Peyer's Patches cells Peyer's Patches cells
  • Lactoferrin acts as a chemokine, a chemokine blocker or a chemokine enhancer
  • 150ul of serum free media containing the chemokine of choice was loaded in the appropriate feeder tray well.
  • the migration chamber was inserted, the cells mixed cells and loaded. Plates were incubated at 37 2 C for 2 hours.
  • the migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 2 C, periodically tilting the plate to help dislodge cells.
  • 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer.
  • FIG. IA shows that lactoferrin can attract as many THP-I cells as MCP-I.
  • TLF lactoferrin.
  • FIG. IB shows that when combined with MCP-I at low doses (0.1 mg/ml), lactoferrin blocks THP-I cell migration towards MCP-I.
  • LF lactoferrin.
  • FIG. 1C shows that when combined with MCP-I at high doses (1 mg/ml), lactoferrin enhances THP-I cell migration towards MCP-I.
  • the migration of different cells and cell lines are tested towards a source of lactoferrin as a chemokine in a chemotaxis or chemoattraction cell-migration assay such as the Chemicon ECM 96-well kit with 175,000 cells per well.
  • the different cells are from human, mouse or other species and include T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
  • T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
  • Jurkat, a human T cell line, and mouse blood cells were tested for chemoattraction to lactoferrin.
  • Blood was collected using anticoagulants (heparinized tubes) and pooled. Volume was 3.5ml for WT (6 mice), 2.5ml for KO (5 mice). One volume of PBS was added.
  • Lympholyte-Mammal (Cedarlane Labs) was gently layered under the diluted blood. Tubes were spun at 800g for 20 minutes and the top layer (serum) was aspirated to a few millimeters above the buffy coat.
  • the buffy coat containing white blood cells (WBC) was collected; next the Lympholyte layer containing granulocytes was collected to just above the pellet containing the red blood cells and dead cells.
  • Samples were brought to 15ml with serum free RPMI and spun again for 10 minutes. Pellets were brought up in 2ml Pharmlyse and incubated for 5 minutes at room temperature in the dark. Samples were diluted to 15ml with serum free RPMI and spun for 10 minutes. Pellets were brought up in 5ml media and counted. 175,000 cells in lOOul volume were loaded per well of a Chemicon ECM515 plate (3 urn pore size) containing 96 wells.
  • 150ul of serum free media containing the chemokine of choice was loaded in the appropriate feeder tray well.
  • the migration chamber was inserted, the cells mixed cells and loaded. Plates were incubated at 37 2 C for 2 hours. The migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 2 C, periodically tilting the plate to help dislodge cells.
  • 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer , gating and counting separately the lymphocytes, granulocytes, macrophages and total cell count for each sample/fraction.
  • FIG. 2A shows that lactoferrin attracts Jurkat cells, a human T cell line, thus functioning as a chemokine for these cells.
  • FIG. 2B shows that lactoferrin attracts mouse granulocytes, thus functioning as a chemokine for these cells.
  • Granulocyes from IL-8 receptor ⁇ knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of granulocytes.
  • FIG. 2C shows that lactoferrin attracts mouse lymphocytes, thus functioning as a chemokine for these cells.
  • Lymphocytes from IL-8 receptor knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of lymphocytes.
  • FIG. 2D shows that lactoferrin attracts mouse macrophages, thus functioning as a chemokine for these cells.
  • Macrophages from IL-8 receptor knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of macrophages.
  • the migration of different cells and cell lines are tested towards a source of chemokine (increasing doses) plus different fixed doses of lactoferrin in a chemotaxis or chemoattraction cell-migration assay such as the Chemicon ECM512 96-well kit with 175,000 cells per well to test blockage or enhancement of cellular migration.
  • the different cells are from human, mouse or other species and include T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
  • APCs antigen presenting cells
  • PBMCs peripheral blood mononuclear cells
  • FIG. 3 shows that lactoferrin binds to human intestinal and fibroblastic cells in a dose-dependent and saturating fashion in the presence of excess of other proteins, thus suggesting specific binding.
  • FIG.3A human intestinal HT-29 cells.
  • FIG. 3B human fibroblasts (NHDF cells) in the presence of 5% BSA.
  • Cells were washed with NaCl/Pi, permeabilized with 0.15% Triton-X-100 in NaCl/Pi for 2 minutes at 2O 0 C, washed again, and blocked with blocking buffer (0.15% Triton X-100, 1% BSA and 3% normal serum in PBS) for 30 minutes at 2O 0 C.
  • the cells were then incubated with primary antibody (mouse monoclonal anti-human lactoferrin; Biodesign #H86024M; 1 :2000 dilution) for 1 hour, followed by biotinylated secondary antibody for 30 minutes, and streptavidin-FITC for 30 minutes. Secondary antibody alone was used as a control to determine specificity.
  • the cells were counterstained with PI and cover-slipped with anti-fade mounting medium. Cells were examined under a fluorescent microscope and digital images taken from each chamber.
  • FIG. 4 shows specific binding of lactoferrin to human intestinal HT-29 cells, fibroblastic NHDF cells and hepatocytic HepG2 cells.
  • Fig. 5 shows specific binding of lactoferrin to dermal cells in the wall of an open wound (5A), which disappears when incubating without the specific antibody (only with the labeling antibody, 5B). Green: anti-LF staining.
  • a full-thickness circular dermal punch wound (10 mm in diameter) was inflicted on the stomach of shaved, anesthetized mice. Care was taken to avoid vessels during injury by elevating the redundant skin and using trans-illumination to avoid vessels. The wounds were then filled with vehicle (PBS) or three different strengths of lactoferrin solution and incubated for five minutes. After washing 5 times with ice-cold saline, the entire wound with 5 mm of adjacent normal tissue was excised to the level of muscle and fat, fixed overnight in 10% neutral buffered formalin, embedded in paraffin, and 5- ⁇ m sections prepared from the wound margin.
  • PBS vehicle
  • lactoferrin solution three different strengths of lactoferrin solution
  • the sections were blocked with normal horse serum and stained with monoclonal mouse anti-lactoferrin antibody (Biodesign) at 1 :20,000 dilution followed by biotinylated horse anti-mouse secondary antibody and Streptavidin-FITC (Vector Laboratories) for detection. Nuclei were counterstained with PI, sections examined under a fluorescent microscope and digital images taken (original magnification x20). Sections without primary antibody served as a negative control.
  • Biodesign monoclonal mouse anti-lactoferrin antibody
  • Streptavidin-FITC Vector Laboratories
  • FIG. 6 shows specific binding to dermal cells in an open wound only when lactoferrin is added to the wound and is absent when PBS is added. Binding increases with increasing doses of lactoferrin. Anti-lactoferrin antibodies are seen in green and nuclei in red. Arrows denote the wound margin; circles denote the dermal portion of the wound wall; (e) keratinocytic epithelium; (d) dermis; (hf) hair follicles; fm (fat and muscle). Original magnification: x20.
  • IC 50 is the concentration causing 50% inhibition and is determined after a range of 9 concentrations of lactoferrin is tested.
  • FIG. 7 shows that lactoferrin binds specifically to receptors of chemotactic, inflammatory and immune mediators such as chemokines in a dose-dependent fashion as determined by the IC 5 Q and Ki.
  • IL-8 beta receptor is involved in neutrophil attraction
  • CCR2 is the receptor for MCP-I
  • CCR3 is a receptor for MCP-2, MCP-3, MCP-3, MIP-Ia, MIP Ib, eotaxin 1-3, and RANTES.
  • MCPs macrophage chemotactic proteins
  • MIPs macrophage inflammatory proteins
  • Example 10 Ability of Iactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors in vivo in the setting of a wound condition
  • FIG. 8 shows that Iactoferrin significantly induced chemokines MIP- l ⁇ and MIP-2 (mouse IL-8), and cytokines IL-6 and TNF- ⁇ in wounds in vivo. Significance was calculated versus placebo at each time point using a 2-tailed Student t test. * P ⁇ 0.05; ** P ⁇ 0.01.
  • FIG. 9 shows that Iactoferrin (rhLF) induced the production of MCP-I (A), IL-8 (B), and IL-6 (C) in a time- and dose-dependent form.
  • rhLF Iactoferrin
  • lactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors: keratinocytes
  • FIG. 10 shows that lactoferrin induces the chemokine MIP-3 ⁇ from keratinocytes in a time and dose dependent fashion. * P ⁇ 0.05; ** P ⁇ 0.01
  • Human hepatocytes were grown in a 96-well format plate, employing conditions established previously for their optimum growth, and then tested for production of chemokine CCL20 (MIP-3 ⁇ ). Four (4) wells per group/conc./time point were used. Cells were exposed to various concentrations of placebo or test compounds: recombinant human lactoferrin from ⁇ Agennix and native lactoferrin purified from human milk (Sigma). Supernatants (75 microL aliquots) were collected from cells at 2 h and 4 h after the administration of test compounds.
  • the cells were washed twice with the medium, then lysed using a Protease Inhibitor Cocktail medium and a homogenizer (e.g., using 20 strokes with a Dounce homogenizer).
  • the cell homogenates was clarified by centrifugation at 1000xg for 20 minutes, supernatant filtered through a 0.22 ⁇ m filter, aliquoted into 100 ⁇ l aliquots, and stored at -80 0 C until testing.
  • FIG. 11 shows that lactoferrin (rhLF) induces the chemokine CCL20 (MIP- 3 ⁇ ) in dose dependent manner at 2 h.
  • HepG2 cells were grown to near confluence (with each well having about 1 MM of cells at confluence). The cells were incubated in vitro with the test compounds at different concentrations and times. At the end of each required period of incubation, supernatants were collected (in two aliquots of 250 ⁇ L) from each well and store frozen for cheraokine analysis. Cells were collected in RNAlater and stored for mRNA analysis by QC-PCR.
  • FIG. 12 shows the effect of various concentrations of lactoferrin at various time points on the protein production for MIP-3- ⁇ (A) and its mRNA message level (B). Induction by lactoferrin of these biomarkers in hepatocytes is both time and dose dependent.
  • Hep-G2 cells Human hepatocytes (Hep-G2 cells) were exposed to various concentrations of lactoferrin or vehicle control for 6 hours. Supernatants were collected and analyzed using RayBiotech, Inc. Human Cytokine Antibody Array.
  • FIG. 13 shows the induction of chemotactic, inflammatory and immune mediators in human hepatocytes as the % change with lactoferrin at 40 mg/niL relative to vehicle treatment.
  • Several inflammatory and immune mediators were increased significantly with lactoferrin treatment, including several chemokines such as MIP-3 ⁇ , IL-8, TECK, I-TAC, Lymphotactin, PARC, MIP- l ⁇ , C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, and Eotaxin-3.
  • chemokines such as MIP-3 ⁇ , IL-8, TECK, I-TAC, Lymphotactin, PARC, MIP- l ⁇ , C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, and Eotax
  • Example 14 Lactoferrin screen to find inhibitors of chemotactic, inflammatory or immune mediators
  • mice Groups of six BALB/c mice were dosed once daily for three consecutive days with oral lactoferrin (2.9 g/m2) or placebo. Mice were injected subcutaneously with a mammary cancer cell line (TUBO) on the third day. On the fourth day, mice were sacrificed and FACS analysis conducted on a single-cell suspension of Peyer's patches isolated from the proximal small intestine. The number of cells in the Peyer's patches increases with lactoferrin relative to placebo.
  • oral lactoferrin 2.9 g/m2
  • TUBO mammary cancer cell line
  • FIG. 14 shows that oral lactoferrin (LF) increases the number of CD4+ T cells, CD8+ T cells, and Natural killer T cells (NKT) in a mouse immune organ (Peyer's patches), suggesting that lactoferrin-induced chemotaxis played a role in the process. * p ⁇ 0.01.
  • Example 16 Modulation of immune cell migration by lactoferrin chemoattraction in vivo
  • Wild type (WT) control mice or IL-8 receptor (IL-8R) knock-out (KO) mice were administered lactoferrin orally: From day 3 to 12 mice (KO and ctrls) were dosed with lactoferrin (LF) at 1000 mg/kg for a total of 8 doses once per day each. On day 13 dosing was terminated and on day 18 mice were sacrificed and the Peyer's Patches were dissected. The Peyer's Patches were then placed in 24-well tissue culture plate in 250 ul RPMI with 10% FBS, 1% PS. Cells were ground up in plate, using the top of a sterile syringe plunger.
  • LF lactoferrin
  • Lactoferrin is directly injected into the Peyer's patch or other types of lymph nodes in mice. At different time points the node is extracted, the number of node cells determined, the percent expression of immune cells determined, and both measurements are compared with nodes injected with vehicle placebo. An anti-lactoferrin antibody is injected together with lactoferrin to neutralize this effect and confirm the specificity of lactoferrin' s chemotactic activity.
  • Example 18 Lactoferrin binding to cells of immune organs with concomitant induction of maturation and/or activation
  • Peyer's patches, mesenteric lymph nodes, splenocytes, and other immune organs are incubated with or without lactoferrin and then analyzed for lactoferrin binding and immune cell markers using FACS analysis.
  • NK natural killer
  • B cells B cells [CD19], and dendritic cells
  • MFI mean fluorescence intensity.
  • FITC green dye.
  • PE red dye.
  • FIG.17 Left Panel: Dermal fibroblasts were incubated with or without Lactoferrin in the presence of 5% bovine serum albumin for 30 min at 37C, stained with a biotinylated rabbit anti-lactoferrin antibody followed by streptavidin-TR, and analyzed using a fluorescent activated cell sorter (FACS) machine.
  • FACS fluorescent activated cell sorter
  • FIG.17 Right Panel: Fibroblasts were cultured in 2% FBS medium for 2 hours at 37 0 C with Lactoferrin, fixed, permeabilized and blocked. The cells were then incubated with a mouse monoclonal anti-human lactoferrin followed by biotinylated secondary antibody and strepavidin-FITC. The cells were counter stained with PI, coverslipped with anti-fade mounting medium, and analyzed by fluorescent microscopy.
  • the migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 oC, periodically tilting the plate to help dislodge cells. 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer.
  • FIG. 18 shows that lactoferrin attracts normal human dermal fibroblasts from adult, thus functioning as a chemokine for these cells.
  • Y axis is the mean chemotactic index defined as the mean number of migrated cells divided by the mean number of cells migrated with medium only (zero lactoferrin).
  • X axis is lactoferrin concentration [micromolar].
  • Example 21 Orally administered lactoferrin localizes to Peyer's patches and binds key immune cells.
  • Lactoferrin binding to cells from the Peyer's patches using flow cytometry Balb/C mice received oral lactoferrin (12.5 mmoles/kg) or vehicle (PBS). Animals were sacrificed at the indicated times and cells from the small intestinal Peyer's patches isolated. Extent of lactoferrin binding was determined by FACS analysis using different gates based on side and forward scatter properties to assess different cell populations. The experiment was performed twice with similar results. Positive control: Peyer's patches cells incubated in vitro with 1 uM lactoferrin. Negative control: untreated Peyer's patches cells (PBS or 0 TLF). Lactoferrin was also quantified in Peyer's patches by ELISA.
  • FIG.19A shows lactoferrin binding to cells from the Peyer's patches using flow cytometry, with a strong signal peaking early between 1 and 15 minutes after oral administration. Forward and side scatter properties of the cells suggested that granulocytes and macrophages are mainly responsible for the binding with lymphocytes mediating a low level of binding as well.
  • FIG 19. B shows quantification of cell lysates by ELISA indicating substantial amounts of lactoferrin in Peyer's patches at 1-15 minutes following oral administration. Both approacnes suggest a range between 0.5-1 uM lactoferrin at the peak, following by degradation at later time points.
  • Example 22 Oral lactoferrin induces migration of dendritic cells to Peyer's patches
  • Lactoferrin was tested for its ability to induce the migration of dendritic cells into the Peyer's patches following oral administration.
  • Mouse dendritic cells DCs
  • DCs Mouse dendritic cells
  • PKH2 Sigma
  • CDl lc+ cells were injected into mice.
  • Lactoferrin 1000 mg/kg was administered by oral gavage starting either the day of the DC injection or 2 days prior to injection and continued for a further two days before animals were sacrificed.
  • Peyer's Patches (5/animal) and spleens were collected and analyzed for labeled cells by flow cytometry. Cells from two or three animals within the same group were pooled to obtain enough cells for analysis.
  • FIG.20A shows that oral lactoferrin induces the migration of labeled dendritic cells into the Peyer's patches compared to untreated mice. Pre-treating the animals with lactoferrin [LF] for two days before the injection of labeled dendritic cells resulted in an enhancement of attraction Jo this organ. Accumulation in Peyer's patches appeared to be specific to this GALT tissue since such a response was not observed in spleen.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention describes the use of chemotactic modulators to treat diseases with a chemotactic, immune or inflammatory compontent. The invention also describes the novel use of lactoferrin to modulate the movement or migration of cells and the immune system through its ability to function as a chemotactic modulator by either directly acting as a chemokine or by modulating chemotactic activity either through the induction or inhibition of other chemotactic modulators or by any other means. The use of this constellation of properties to treat diseases with a chemotactic, immune or inflammatory component is envisaged.

Description

USE OF LACTOFERRIN AS A CHEMOKINE AND A CHEMOTACTIC MODULATOR
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to Provisional Application No. 60/709,735 filed on August 19, 2005
TECHNICAL FIELD
[0002] The present invention describes the use of chemotactic modulators to treat diseases with a chemotactic, immune or inflammatory component. The invention also describes the novel use of lactoferrin to modulate the movement or migration of cells and the immune system through its ability to function as a chemotactic modulator by either directly acting as a chemokine or by modulating chemotactic activity either through the induction or inhibition of other chemotactic modulators or by any other means. The use of this constellation of properties to treat diseases with a chemotactic, immune or inflammatory component is envisaged.
BACKGROUND OF THE INVENTION
[0003] With the advancement of scientific knowledge, it has become evident that several diseases or conditions have an inflammatory or immunological component where a complex network of cytokines, chemokines, and growth factors is present, modulating the migration and/or activation of different inflammatory or immune cells.
[0004] Lactoferrin is a single chain metal binding glycoprotein. Many cells types, such as epithelial cells, monocytes, macrophages, lymphocytes, and intestinal brush-border cells, are known to have lactoferrin receptors. In addition to lactoferrin being an essential growth factor for both B and T lymphocytes, lactoferrin has a wide array of functions related to host primary defense mechanisms. For example, lactoferrin has been reported to activate natural killer (NK) cells, induce colony-stimulating activity, activate polymorphonuclear neutrophils (PMN), regulate granulopoeisis, enhance antibody-dependent cell cytotoxicity, stimulate lymphokine-activated killer (LAK) cell activity, and potentiate macrophage toxicity.
[0005] What has not been hitherto described is lactoferrin' s ability to modulate the chemotactic milieu in a unique fashion. BRIEF SUMMARY OF THE INVENTION
[0006] The present invention is directed to a method for modulating the migration and chemokine production of cells by administration of a lactoferrin preparation which may be applied topically, orally or parenterally for treatment of diseases with an immune or inflammatory component. In vitro, the method involves screening for agents that will inhibit the secretion of chemokines and cytokines or the chemotaxis toward a lactoferrin gradient.
[0007] The following numbered sentences more readily define the invention as described herein.
1. A method of treating a disease or condition in a subject comprising the step of administering to the subject a chemotactic modulator composition in an amount sufficient to modulate chemotactic activity in said subject.
2. The method of sentence 1, wherein the disease or condition is selected from the group consisting of a hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
3. The method of sentence 1, wherein chemotactic^ modulator is selected from the group consisting of lactoferrin, chemokine, chemokine receptor, inflammatory or immune mediator, viral anti-chemokine, a mixture of casein and autologous serum, chemotactic C5 fragment, formyl-1-methionyl-l-leucine-l-phenylalanine (f-Met-Leu-Phe), bacterial factor derived from Escherichia coli, the simple peptide formylmethionylalanine, and activated human complement, carbamylcholine, phenylephrine, and cyclic guanosine 5'- monophosphate, nucleosides and nucleotides and prostaglandins
4. A method of treating a disease or condition in a subject comprising the step of administering to a subject a lactoferrin composition in an amount sufficient to modulate chemotactic activity in said subject.
5. The method of sentence 4, wherein modulation refers to increase.
6. The method of sentence 4, wherein modulation refers to decrease.
7. The method of sentence 4, wherein chemotactic activity refers to attraction of cells by lactoferrin' s ability to act as a chemokine. 8. The method of sentence 4, wherein chemotactic activity refers to an activity that influences the movement or migration of cells either through the induction or inhibition of other chemotactic modulators.
9. The method of sentence 4, wherein said lactoferrin composition is dispersed in a pharmaceutically acceptable carrier.
10. The method of sentence 4, wherein said lactoferrin is mammalian lactoferrin.
11. The method of sentence 4, wherein said lactoferrin is human or bovine.
12. The method of sentence 4, wherein said lactoferrin is recombinant lactoferrin.
13. The method of sentence 4, wherein said lactoferrin is produced by purification from natural sources or artificially.
14. The method of sentence 4, wherein said lactoferrin composition comprises an amino acid sequence selected from the group consisting of SEQ.ID.NO.l, SEQ.ID.NO.2, SEQ.ID.NO.3, SEQ.ID.NO.4, SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.il.
15. The method of sentence 4, wherein said lactoferrin is administered topically, orally or parenterally.
16. The method of sentence 4, wherein the amount of the composition that is administered is about lμg to about lOOg per day.
17. The method of sentence 4, wherein said lactoferrin composition is a topical gel, a solution, or a tablet containing a concentration of about 0.01% to about 20% of lactoferrin.
18. The method of sentence 17, wherein said topical gel is composed from the polymers selected from the group of carbopol, carboxymethyl-cellulose, and pluronic polymers.
19. The method of sentence 4, wherein the disease or condition are selected from the group consisting of hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
20. The method of sentence 19, wherein the skin disease is selected from the group consisting of psoriasis, scleroderma, systemic lupus erythematosus, atopic dermatitis, epidermolysis bullosa, acute wounds, chronic wounds, and burns.
21. The method of sentence 19, wherein the gastrointestinal disorder is Crohn's disease, colitis, necrotizing entercolitis, endometriosis, irritable bowel syndrome, pancreatitis, periodontal disease, and ulcerative colitis. 22. The method of sentence 19, wherein the fibrotic disease is selected from the group consisting of diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis, and atherosclerosis.
23. The method of sentence 19, wherein the inflammatory condition is of an infectious origin or a non-infectious origin.
24. The method of sentence 23, wherein the infectious inflammatory disease is casued by a pathogen selected from the group consisting of meningococci, Salmonella typhi, Dengue virus, Plasmodia, Pneumocystis carinii, Yersinia enterocolitica pathogens, and enterovirus, fungi.
25. The method of sentence 23, wherein non-infectious inflammatory disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ-specific inflammatory diseases, glomerular disease, Sjogrens disease, Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
26. The method of sentence 19, wherein the wound condition is selected from the group consisting of chronic wound, acute wound, skin wound, internal wound, gastrointestinal wound, oral wound, ophthalmic wound, surgical wound, incisional wound, excisional wound, diabetic ulcer, venous stasis ulcer, decubitus or pressure ulcer, chemical wound, burn wound, full-thickness skin wound and partial-thickness skin wound.
27. The method of sentence 19, wherein the autoimmune disease is selected from the group consisting of Active Chronic Hepatitis, Addison's Disease, Anti-phospholipid Syndrome, Atopic Allergy, Atrophic Gastritis, Achlorhydra Autoimmune, Celiac Disease, Crohn's Disease, Cushings Syndrome, Dermatomyositis, (Type I) Diabetes, Discoid Lupus, Erythematosis, Goodpasture's Syndrome, Grave's Disease, Hashimoto's Thyroiditis, Idiopathic Adrenal Atrophy, Idiopathic Thrombocytopenia, Insulin-dependent Diabetes, Lambert-Eaton Syndrome, Lupoid Hepatitis, Lymphopenia, Mixed Connective Tissue Disease, Multiple Sclerosis, Pemphigoid, Pemphigus Vulgaris, Pernicious Anema, Phacogenic Uveitis, Polyarteritis Nodosa, Polyglandular Auto Syndromes, Primary Biliary Cirrhosis, Primary Sclerosing Cholangitis, Psoriasis, Raynauds, Reiter's Syndrome, Relapsing Polychondritis, Rheumatoid Arthritis, Schmidt's Syndrome, Scleroderma-CREST, Sjogren's Syndrome, Sympathetic Ophthalmia, Systemic Lupus Erythematosis, Takayasu's Arteritis, Temporal Arteritis, Thyrotoxicosis, Type B Insulin Resistance, Ulcerative Colitis, Wegener's Granulomatosis, graft versus host disease (GVHD), organ transplant rejection, autoimmune hepatitis, autoimmune cholangitis, irritable bowel syndrome (IBS), chronic granulomatous disease, ankylosing spondylitis, scleroderma, polymyositis, (dermato)myositis, and systemic vasculitis.
28. The method of sentence 19, wherein the hyperproliferative disease is further defined as cancer.
29. The method of sentence 28, wherein the cancer comprises a neoplasm.
30. The method of sentence 29, wherein the neoplasm is selected from the group consisting of melanoma, non-small cell lung, small-cell lung, lung hepatocarcinoma, retinoblastoma, astrocytoma, gliobastoma, leukemia, neuroblastoma, squamous cell, head, neck, gum, tongue, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, sarcoma, cervical, gastrointestinal, lymphoma, brain, colon, and bladder.
31. The method of sentence 29, wherein the neoplasm is a hematopoietic neoplasm.
32. The method of sentence 30, wherein the hematopoietic neoplasm is selected from the group consisting of acute myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, and chronic lymphocytic leukemia.
33. The method of sentence 19, wherein the hyperproliferative disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, osteoarthritis, leiomyomas, adenomas, lipomas, hemangiomas, fibromas, vascular occlusion, restenosis, atherosclerosis, pre-neoplastic lesions, carcinoma in situ, oral hairy leukoplakia, and psoriasis.
34. The method of sentence 19, wherein the endocrine and/or metabolism disorder is diabetes mellitus, thyroid disorder, or fragile bone disease.
35. The method of sentence 33, wherein the fragile bone disease is selected from the group consisting of osetoporosis, osteitis deformans, osteogenesis imperfecta, osteopetrosis, Rickets, and osteomalacia.
36. The method of sentence 19, wherein the pain is acute or chronic pain.
37. The method of sentence 19, wherein the cardiovascular disease is congestive heart failure, hypertension, cardiomyopathy, myocarditis, atherosclerosis, chronic venous disease, or heart arrhythmia.
38. The method of sentence 19, wherein the hematological disorder is anaemia, sickle cell anemia or cachexia. 39. The method of sentence 19, wherein the ocular disorder is conjunctivitis, dry eye disease, glaucoma, allergic eye disease, uveitis or ocular infection.
40. The method of sentence 19, wherein the respiratory disorder is atopic asthma, non-atopic asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis, allergic rhinitis, fibrotic lung disease, COPD, ARDS, pulmonary vascular disease/pulmonary hypertension, Cor Pulmonale, or cystic fibrosis.
41. The method of sentence 4, further comprising administering a subject a lactoferrin composition in combination with other appropriate therapy.
42. A method of treating a disease or condition in a subject by administering to the subject a sufficient amount of a lactoferrin composition, wherein the lactoferrin composition binds to receptors of chemotactic, immune, or inflammatory mediators.
43. The method of sentence 42, wherein the chemotactic, immune or inflammatory mediators are chemokines and other chemotactic modulators, cytokines, or growth factors.
44. The method of sentence 42, wherein disease or condition are selected from the group consisting of skin disease, fibrotic disease, chronic inflammatory disease, wound condition, inflammatory condition, autoimmune disease, hyperproliferative disease, fragile bone disease, pain and diabetes mellitus.
45. The method of sentence 42, wherein lactoferrin acts directly as a chemokine to attract cells.
46. The method of sentence 42, wherein the binding of lactoferrin to the receptors inhibits the chemotactic, immune or inflammatory mediators.
47. The method of sentence 42, wherein the binding of lactoferrin to the receptors stimulates the chemotactic, immune or inflammatory mediators.
48. The method of sentence 42, wherein the chemokines are MIP-I alpha, MIP-I β, IL-8, MIP-2,
MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha, TECK, I-TAC, Lymphotactin, PARC, MIP-ldelta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO-α, GRO-β, GRO-γ, SCF, or MEC/CCL28.
49. The method of sentence 42, wherein the receptors are IL-8 alpha receptor, IL-8 beta receptor, neuropeptide Yl receptor, PACAP receptor, K+ channel receptor, CCR2, CCR3, CCR6, CXCR4, EGFR, TLR-2, TLR-3, TLR-4, TLR4/MD, Mannose receptor, CD 14, or TRAIL R4.
50. A method of stimulating a cultured cell to induce chemotactic, inflammatory and/or immune mediators comprising the step of adding a lactoferrin composition to the cell thereby stimulating MIP-lalpha, MlP-lβ, IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP- 3alpha, TECK5 I-TAC, Lymphotactin, PARC, MIP-I delta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO-α, GRO-β, GRO-γ, SCF5 or MEC/CCL28.
[0008] The foregoing has outlined rather broadly the features and technical advantages of the present invention in order that the detailed description of the invention that follows may be better understood. Additional features and advantages of the invention will be described hereinafter which form the subject of the claims of the invention. It should be appreciated by those skilled in the art that the conception and specific embodiment disclosed may be readily utilized as a basis for modifying or designing other structures for carrying out the same purposes of the present invention. It should also be realized by those skilled in the art that such equivalent constructions do not depart from the spirit and scope of the invention as set forth in the appended claims. The novel features which are believed to be characteristic of the invention, both as to its organization and method of operation, together with further objects and advantages will be better understood from the following description when considered in connection with the accompanying figures. It is to be expressly understood, however, that each of the figures is provided for the purpose of illustration and description only and is not intended as a definition of the limits of the present invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0009] For a more complete understanding of the present invention, reference is now made to the following descriptions taken in conjunction with the accompanying drawing, in which:
[0010] FIG. 1 shows lactoferrin acting as a chemokine, a chemokine activity blocker, and a chemokine activity enhancer.
[0011] FIG. 2 shows attraction of different cell types to lactoferrin, a process that is in part mediated by the IL- 8 receptor.
[0012] FIG. 3 shows specific binding of lactoferrin to different cell types as assessed by flow cytometry. [0013] FIG. 4 shows specific binding of lactoferrin to different cell types as assessed by fluorescent microscopy.
[0014] FIG. 5 shows specific binding of lactoferrin to deπnal cells in vivo.
[0015] FIG. 6 shows a dose response of lactoferrin binding to dermal cells in vivo.
[0016] FIG. 7 shows specific binding of lactoferrin to receptors for inflammatory or immune mediators.
[0017] FIG. 8 shows that topical lactoferrin activates the biological activity of inflammatory or immune receptors in vivo in the setting of an open wound.
[0018] FIG. 9 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human fibroblasts.
[00191 FIG. 10 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human keratinocytes.
[0020] FIG. 11 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in human hepatocytes.
[0021] FIG. 12 shows that lactoferrin activates the biological activity of inflammatory or immune receptors in hepatocytes in a time and dose dependent fashion.
[0022] FIG. 13 shows the release of different inflammatory or immune mediators form hepatocytes upon lactoferrin stimulation.
[0023] FIG. 14 shows that oral lactoferrin increases the percentage of different immune cells in an immune organ (Peyer's patches) of mice, suggesting that lactoferrin-induced chemotaxis played a role in the process. [0024] FIG. 15 shows that oral lactoferrin increases the total number of cells in a mouse immune organ (Peyer's patches), a process that is significantly impaired in IL-8 receptor knockout mice, suggesting that lactoferrin-induced chemoattraction played a role and that this chemotaxis is mediated at least in part by the IL-8 receptor in vivo.
[0025] FIG. 16 shows that lactoferrin binds to all tested immune cells of a mouse immune organ (spleen), inducing concomitant maturation and/or activation in some cell types.
[0026] FIG. 17. shows the ability of lactoferrin to bind fibroblasts in vitro by two methods.
[0027] FIG. 18 shows that lactoferrin attracts normal human dermal fibroblasts from adult, thus functioning as a cheniokine for these cells.
[0028] FIG.19. shows lactoferrin binding to cells from mouse Peyer's patches with a strong signal peaking early between 1 and 15 minutes after oral administration.
[0029] FIG. 20. shows that oral lactoferrin induces the migration of labeled dendritic cells into the Peyer's patches
DETAILED DESCRIPTION OF THE INVENTION
[0030] It is readily apparent to one skilled in the art that various embodiments and modifications can be made to the invention disclosed in this Application without departing from the scope and spirit of the invention.
[0031] As used herein, the use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." Still further, the terms "having", "including", "containing" and "comprising" are interchangeable and one of skill in the art is cognizant that these terms are open ended terms. [0032] The term "acute pain" as used herein includes tissue injury or pathology. For example, acute pain includes, but is not limited to pain following surgery (e.g., surgery), pain following trauma (e.g., blunt or sharp injury, bullet wounds), pain associated with athletic injury, and the occasional headache. Medically, acute pain can have a diagnostic value; that is, it leads to the discovery of a pathological condition. Yet further, acute pain lasts or is anticipated to last for a short period of time, typically less than one to two months. Acute pain is associated with hyperactivity of the sympathetic nervous system, for example, tachycardia, increased respiratory rate, increased blood pressure, increased Cortisol release, diaphoresis and dilated pupils.
[0033] The term "antibiotic" as used herein is defined as a substance that inhibits the growth of microorganisms without damage to the host. For example, the antibiotic may inhibit cell wall synthesis, protein synthesis, nucleic acid synthesis, or alter cell membrane function. Classes of antibiotics that can possibly be used include, but are not limited to, macrolides (e.g., erythromycin), penicillins (e.g., nafcillin), cephalosporins (e.g., cefazolin), carbepenems (e.g., imipenem, aztreonam), other beta-lactam antibiotics, beta-lactam inhibitors (e.g., sulbactam), oxalines (i.e. linezolid), aminoglycosides (e.g., gentamicin), chloramphenicol, sufonamides (e.g., sulfamethoxazole), glycopeptides (e.g., vancomycin), quinolones (e.g., ciprofloxacin), tetracyclines (e.g., minocycline), fusidic acid, trimethoprim, metronidazole, clindamycin, mupirocin, rifamycins (e.g., rifampin), streptogramins (e.g., quinupristin and dalfopristin) lipoprotein (e.g., daptomycin), polyenes (e.g., amphotericin B), azoles (e.g., fluconazole), and echinocandins (e.g., caspofungin acetate). The term "morbidity" as used herein is the state or condition of being diseased. Yet further, morbidity can also refer to the ratio of incidence, for example the number of sick subjects or cases of diseases in relationship to a specific population.
[0034] The term "anti-depressant drug" as used herein includes compounds that provide pain relief by reducing side-effects including anxiety. Anti-depressant drugs include, for example, but not limited to tricyclin antidepressants, sedatives, tranquilizers, hypnotics, antihistamines, and amphetamines.
[0035] The term "antimicrobial" as used herein is defined as a substance that inhibits the growth of microorganisms without damage to the host, for example antibiotics, antifungal and antiseptics. [0036] The term "atherosclerosis" as used herein includes a form of arteriosclerosis characterized by a combination of changes in the intima of arteries, such changes include, but are not limited to accumulation of lipids, complex carbohydrates, blood and blood products, fibrous tissue and calcium deposits. Yet further, atherosclerotic plaques can be characterized into at least two areas. One type is characterized by prominent proliferation of cells with small accumulations of lipids. The second type consists mostly of intracellular and extracellular lipid accumulation and a small amount of cellular proliferation.
[0037] The term "bacteremia" as used herein is defined as having a focus of bacterial infection or bacteria in the blood of the subject.
[0038] The term "cancer" as used herein is defined as a hyperproliferation of cells whose unique trait — loss of normal controls — results in unregulated growth, lack of differentiation, local tissue invasion, and metastasis. Examples include but are not limited to, melanoma, non-small cell lung, small-cell lung, lung, hepatocarcinoma, leukemia, retinoblastoma, astrocytoma, glioblastoma, gum, tongue, neuroblastoma, head, neck, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, cervical, gastrointestinal, lymphoma, brain, colon, sarcoma or bladder.
[0039] The term "cardiovascular disease or disorder" as used herein refers to disease and disorders related to the cardiovascular or circulatory system. Cardiovascular disease and/or disorders include, but are not limited to, diseases and/or disorders of the pericardium, heart valves (e.g., incompetent valves, stenosed valves, rheumatic heart disease, mitral valve prolapse, aortic regurgitation), myocardium (e.g., coronary artery disease, myocardial infarction, heart failure, ischemic heart disease, angina) blood vessels (e.g., hypertension, arteriosclerosis, aneurysm) or veins (e.g., varicose veins, hemorrhoids). Yet further, one skill in the art recognizes that cardiovascular diseases and/or disorders can result from congenital defects, genetic defects, environmental influences (e.g., dietary influences, lifestyle, stress, etc.), and other defects or influences.
[0040] The term "cell," "cell line," and "cell culture" as used herein may be used interchangeably. All of these terms also include their progeny, which are any and all subsequent generations. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations.
[0041] The term "cytokine" as used herein refers to proteins that are made by cells that affect the behavior of other cells, for example stimulate or inhibit cell proliferation. For example, cytokines that are made by lymphocytes are often called lymphokines or interleukins. One of skill in the art realizes that the term cytokine is a generic term used in the literature to refer to proteins that are made by cells that can effect the behavior of other cells.
[0042] The term "chemokine" as used herein refers to small cytokines that are involved in the migration and activation of cells, for example phagocytic cells and lymphocytes. One of skill in the art realizes that chemokines play a central role in inflammatory and immune response processes.
[0043] The term "cholesterol" as used herein refers to the monohydric alcohol form, which is a white, powdery substance that is found in all animal cells and in animal-based foods (not in plants). Cholesterol is an essential nutrient necessary for many functions, including the following: repairing cell membranes, manufacturing vitamin D on the skin's surface, production of hormones, such as estrogen and testosterone, and possibly helping cell connections in the brain that are important for learning and memory.
[0044] The term "chronic pain" as used herein refers to pain that lasts longer than 1 month or beyond the resolution of an acute tissue injury or is recurring or is associated with tissue injury and/or chronic diseases that are expected to continue or progress, for example, cancer, arthritis, inflammatory disease, chronic wounds, cardiovascular accidents, spinal cord disorders, central nervous system disorder or recovery from surgery. Chronic pain can be associated with several factors that include, but are not limited to lassitude, sleep disturbance, decreased appetite, loss of taste for food, weight loss, diminished libido, constipation, or depression.
[0045] The term "chylomicron" as used herein refers to the largest in size and lowest in density of the triglyceride carrying lipoproteins. [0046] The term "diabetes mellitus" as used herein refers to a disorder of carbohydrate metabolism that is typically characterized by hyperglycemia and glycosuria, which results from inadequate production or utilization of insulin. Diabetes mellitus includes several syndromes or disorders, for example, but not limited to, primary diabetes mellitus (e.g., insulin- dependent (Type I) and non-insulin-dependent (Type II)); secondary diabetes [for example: pancreatic diabetes (e.g., destruction of the pancreas, removal of the pancreas, etc.); extrapancreatic/endocrine diabetes (e.g., hypersomatotropism, hyperadrenalism, hyperthyroidism, glucagonama, etc.); drug-induced diabetes (e.g., steroid diabetes, thiazides, etc.)] and rare/exceptional forms of diabetes (e.g., lipoatrophic diabetes, myatonic diabetes, disturbance of insulin receptors, genetic syndromes, etc). Long-term complications of diabetes include neuropathy, retinopathy, nephropathy, generalized degenerative changes in the blood vessels and increased susceptibility to infection.
[0047] The term "effective amount" or "therapeutically effective amount" as used herein refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
[0048] The term "lactoferrin" or "LF" as used herein refers to native or recombinant lactoferrin. Native lactoferrin can be obtained T>y purification from mammalian milk or colostrum or from other natural sources. Recombinant lactoferrin (rLF) can be made by recombinant expression or direct production in genetically altered animals, plants, fungi, bacteria, or other prokaryotic or eukaryotic species, or through chemical synthesis.
[0049] The term "lactoferrin composition" as used herein refers to a composition having lactoferrin or a part thereof, wherein at least the N-terminal glycine residue is truncated or substituted.
[0050] The term "N-terminal lactoferrin variant" as used herein refers to lactoferrin wherein at least the N-terminal glycine has been truncated and/or substituted. N-terminal lactoferrin variants also include, but are not limited to deletion and/or substitution of one or more N-terminal amino acid residues, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 N-terminal amino acid residues, etc. Thus, N-terminal lactoferrin variants comprise at least deletions or truncations and/or substitutions of 1 to 16 N-terminal amino acid residues. The deletion and/or substitution of at least the N-terminal glycine of lactoferrin mediates the same biological effects as full-length lactoferrin and/or may enhance lactoferrin' s biological activity, for example by stimulating the production of various cytokines (i.e., IL-18, MIP-3α, GM-CSF or IFN-γ by inhibiting various cytokines, (i.e., IL-2, IL-4, IL-5, IL-IO, and TNF-α and/or by stimulating.
[0051] The term "fragile bone condition" or "fragile bone disease" as used herein refers to a condition or disease characterized by low bone mass or structural deterioration of bone tissue, leading to bone fragility and increased susceptibility to fractures.
[0052] The term "gastrointestinal disorder or conditions" as used herein includes gastrointestinal disorders in which one or more of the symptoms and conditions affect the gastrointestinal tract from the mouth to the anus, as well as any organ that may play a role in digestion, for example, but not limited to pancreas, or gall bladder. Gastrointestinal disorders include, but are not limited to, heartburn, bloating, postoperative ileus, abdominal pain and discomfort, early satiety, epigastric pain, nausea, vomiting, burbulence, regurgitation, intestinal pseudoobstruction, anal incontinence, gastroesophageal reflux disease, irritable bowel syndrome, ulcerative colitis, Crohn's disease, renal disorders, menstrual cramps, pancreatitis, spastic and interstitial cystitis and ulcers and the visceral pain associated therewith.
[0053] The term "Grade 3 neutropenia" refers to the reduction of the absolute neutrophil cell count (ANC) to less than about 1000 cells/μL. The term "neutropenia" generally refers to a condition in which the ANC is reduced to 1000 cells//μL or less. Such a condition may be caused by depressed production, increased peripheral destruction of neutrophils. The most common neutropenias are iatrogenic, resulting from the widespread use of cytotoxic or immunosuppressive therapies for cancer treatment or control of autoimmune disorders. Other causes of neutropenia include induction by drugs, hematological diseases including idiopathic, cyclic neutropenia, Chediak-Higashi syndrome, aplastic anemia, infantile genetic disorders, tumor invasion such as myelofibrosis, nutritional deficiency; infections such as tuberculosis, typhoid fever, brucelloisis, tularemia, measles, infectious mononucleosis, malaria, viral hepatitis, leishmaniasis, AIDS, antineutrophil antibodies and/or splenetic or lung trapping, autoimmune disorders, wegner's granulomatosis, acute endotoxemia, hemodialysis, and cardiopulmonary bypass. The present invention applies to any acquired and inherited neutropenic conditions. [0054] The term "graft-versus-host-disease" or "GVHD" as used herein is the pathological reaction that occurs between the host and grafted tissue. The grafted or donor tissue dominates the pathological reaction. Graft-versus-host-disease (GVHD) can be seen following stem cell and/or solid organ transplantation. GVHD occurs in immunocompromised subjects, who when transplanted, receive "passenger" lymphocytes in the transplanted stem cells or solid organ. These lymphocytes recognize the recipient's tissue as foreign. Thus, they attack and mount an inflammatory and destructive response in the recipient. GVHD has a predilection for epithelial tissues, especially skin, liver, and mucosa of the gastrointestinal tract. GVHD subjects are immunocompromised due the fact that prior to transplant of the graft, the subject receives immunosuppressive therapy.
[0055] The term "gram-negative bacteria" or "gram-negative bacterium" as used herein is defined as bacteria which have been classified by the Gram stain as having a red stain. Gram-negative bacteria have thin walled cell membranes consisting of a single layer of peptidoglycan and an outer layer of lipopolysaccharide, lipoprotein, and phospholipid. Exemplary organisms include, but are not limited to, Enterobacteriacea consisting of Escherichia, Shigella, Edwardsiella, Salmonella, Citrobacter, Klebsiella, Enterobacter, Hafnia, Serratia, Proteus, Morganella, Providencia, Yersinia, Erwinia, Buttlauxella, Cedecea, Ewingella, Kluyvera, Tatumella and Rahnella. Other exemplary gram-negative organisms not in the family Enterobacteriacea include, but are not limited to, Pseudomonas aeruginosa, Stenotrophomonas maltophilia, Burkholderia, Cepacia, Gardenerella, Vaginalis, and Acinetobacter species.
[0056] The term "gram-positive bacteria" or "gram-positive bacterium" as used herein refers to bacteria, which have been classified using the Gram stain as having a blue stain. Gram-positive bacteria have a thick cell membrane consisting of multiple layers of peptidoglycan and an outside layer of teichoic acid. Exemplary organisms include, but are not limited to, Staphylococcus aureus, coagulase-negative staphylococci, streptococci, enterococci, corynebacteria, and Bacillus species.
[0057] The term "high-density lipoprotein" or "HDL" as used herein is the smallest and most dense type of cholesterol-carrying lipoprotein and is often referred to as the "good" cholesterol. [0058] The term "hyperproliferative disease" is defined as a disease that results from a hyperproliferation of cells. Exemplary hyperproliferative diseases include, but are not limited to cancer or autoimmune diseases. Other hyperproliferative diseases may include vascular occulsion, restenosis, atherosclerosis, or inflammatory bowel disease.
[0059] The term "immunocompromised" as used herein is defined as a subject who is, at the time of pathogen exposure, has a pre-existing condition that reduces one or more mechanisms for normal defense against infection. The immunocompromised condition may be due to a defect or dysfunction of the immune system or to other factors that heighten susceptibility to infection, for example immunosuppressive agents. Although such a categorization allows a conceptual basis for evaluation, immunocompromised individuals with infection often do not fit completely into one group or the other. More than one defect in the body's defense mechanisms may be affected. For example, an immunocompromised state can result from indwelling central lines or other types of impairment due to intravenous drug abuse; or be caused by secondary malignancy, malnutrition, or having been infected with other infectious agents such as tuberculosis, influenza, Staphylococcus aureus or sexually transmitted diseases, e.g., syphilis or hepatitis.
[0060] The term "insulin-dependent diabetes mellitus"; "IDDM" or "Type I" refer to diabetes that is characterized by a hyperglycemia, glycosuria, and low blood insulin levels. Type I diabetes can develop at any age. It typically has an abrupt onset during the first two decades of life. Insulin therapy is usually required.
[0061] The term "intermediate density lipoprotein" or "IDL" as used herein refers to a triglyceride-carrying lipoprotein.
[0062] The term "lipid" as used herein refers to the building blocks of any of the fats or fatty substances found in animals and plants, which are characterized by their insolubility in water and solubility in fat solvents such as alcohol, ether and chloroform. Lipids include fats (e.g., esters of fatty acids and glycerol); lipoids (e.g., phospholipids, cerebrosides, waxes) and sterols (e.g., cholesterol). [0063] The term "lipoproteins" as used herein are protein spheres that transport cholesterol, triglyceride, or other lipid molecules through the bloodstream. Lipoproteins are categorized into five types according to size and density. They can be further defined by whether they carry cholesterol [the two smaller lipoproteins (HDL and LDL)] or triglycerides [the three largest lipoproteins (IDL, VLDL, and chylomicrons)].
[0064] The term "low density lipoprotein" or "LDL" as used herein is a type of cholesterol-carrying lipoprotein which is often called the "bad" cholesterol.
[0065] The term "metal chelator" as used herein refers to a compound which binds metal. Metal chelators that can be used in the present invention include the divalent metal chelators, for example, ethylenediaminetetraacetic acid (EDTA), [ethylenebis (oxyethylenenitrilo)] tetraacetic acid (EGTA), l,2-bis(2-aminophenoxy)ethane-N,N,N',N'- tetraacetic acid (BAPTA), hydroxyethlene triamine diacetic acid, (HEDTA) or salts thereof.
[0066] The term "morbidity" as used herein is the state of being diseased. Yet further, morbidity can also refer to the disease rate or the ratio of sick subjects or cases of disease in to a given population.
[0067] The term "mortality" as used herein is the state of being mortal or causing death. Yet further, mortality can also refer to the death rate or the ratio of number of deaths to a given population.
[0068] The term "neurology" or "neurological" refers to conditions, disorders, and/or diseases that are associated with the nervous system. Thus, any condition, disorder and/or disease that effects any component or aspect of the nervous system (either central or peripheral) is referred to as a neurological condition, disorder and/or disease. As used herein, the term "neurological" or "neurology" encompasses the terms "neuropsychiatric" or "neuropsychiatry" and "neuropsychological" or "neuropsychological". Thus, a neurological disease, condition, or disorder includes, but is not limited to cognitive disorders, affective disorders (e.g., depression disorders and/or anxiety disorders), movement disorders, mental disorders, pain disorders, sleep disorders, etc. [0069] The term "neutropenia" as used herein refers to an a decrease or small number of neutrophils in the blood compared to normal. For example, the World Health Organization defines neutropenia as a subject having an absolute neutrophil cell count (ANC) of about 2000 cells/μL or less. Thus, as used herein a subject suffering from neutropenia is one having an ANC of about 2000 cells/μL or less, for example 1000 cells/μL or even less than 500 cells/μL.
[0070] The term "non-insulin-dependent diabetes mellitus"; "NIDDM" or Type II" refer to diabetes that is characterized by hyperglycemia and insulin levels being normal to high. Typically, Type II diabetes is a form of diabetes mellitus that has gradual onset in obese individuals over the age of 35. Insulin therapy is usually not required; however, Type II diabetes can be destined or prone to become fully insulin-dependent.
[0071] The term "organ or tissue transplant rejection" as used herein refers to a consequence of organ or tissue transplantation caused by the recipient's or host's immune system in response to the transplanted organ/tissue, which can damage or destroy it. Thus, one of skill in the art realizes that "organ or tissue transplant rejection" is controlled by the host subject.
[0072] The term "osteoporosis" as used herein is defined as a general term for describing any disease process that results in reduction in the mass of bone.
[0073] The term "pain" as used herein refers to an unpleasant sensation. For example, the subject experiences discomfort, distress or suffering. Pain of a moderate or high intensity is typically accompanied by anxiety. Thus, one of skill in the art is cognizant that pain may have dual properties, for example sensation and emotion.
[0074] The term "parenteral administration" as used herein includes any form of administration in which the compound is absorbed into the subject without involving absorption via the intestines. Exemplary parenteral administrations that are used in the present invention include, but are not limited to intramuscular, intravenous, intraperitoneal, intraoccular, or intraarticular administration. Yet further, parenteral administration also includes administration into a surgical field. [0075] The term "pharmaceutically acceptable carrier" as used herein includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the vectors or cells of the present invention, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
[0076] The term "pharmaceutical composition" as used herein refers to a lactoferrin composition that this dispersed in a pharmaceutically acceptable carrier. The lactoferrin composition can comprise lactoferrin or an N-terminal lactoferrin variant in which at least the N-terminal glycine amino acid residue is truncated or substituted.
[0077] The term "oral administration" as used herein includes, but is not limited to oral, buccal, enteral or intragastric administration.
[0078] The term "subject" as used herein, is taken to mean any mammalian subject to which a human lactoferrin composition is orally administered according to the methods described herein. In a specific embodiment, the methods of the present invention are employed to treat a human subject. Another embodiment includes treating a human subject suffering from a wound.
[0079] The term "therapeutically effective amount" as used herein refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
[0080] The term "topical administration" as used herein includes, but is not limited to topical, dermal (e.g., trans-dermal or intra-dermal), epidermal, or subcutaneous.
[0081] The term "treating" and "treatment" as used herein refers to administering to a subject a therapeutically effective amount of a recombinant human lactoferrin composition so that the subject has an improvement in the disease. The improvement is any improvement or remediation of the symptoms. The improvement is an observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the disease condition, but may not be a complete cure for the disease.
[0082] The term "wound" as used herein refers to any injury, such as an ulcer, as a result of disease or disorder, or as a result of an accident, incident, or surgical procedure. Wound can be further defined as acute and/or chronic.
[0083] The term "preventing" as used herein refers to minimizing, reducing or suppressing the risk of developing a disease state or parameters relating to the disease state or progression or other abnormal or deleterious conditions.
[0084] The term "respiratory disorder" refers to any condition and/or disorder relating to respiration and/or the respiratory system. The respiratory disorder can be an allergic or non-allergic respiratory disorder. More specifically, the respiratory disorder includes, but is not limited to atopic asthma, non-atopic asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis and allergic rhinitis.
[0085] The term "sepsis" as used herein is defined as a Systemic Inflammatory Response Syndrome to an infective process in which severe derangement of the host immune system fails to prevent extensive 'spill over' of inflammatory mediators from a local infection focus into the systemic circulation.
[0086] The term "septic shock" as used herein is a consequence of sepsis in which the systemic inflammatory response leads to the failure of vital organs' function (for example of the lungs as in ARDS).
[0087] The term "subject" as used herein, is taken to mean any mammalian subject to which a lactoferrin composition is administered according to the methods described herein. Thus, a skilled artisan realizes that a mammalian subject, includes, but is not limited to humans, monkeys, horses, pigs, cows, dogs, cats, rats and mice. In a specific embodiment, the methods of the present invention are employed to treat a human subject. [0088] The term "Th2 cells" as used herein is defined as a subset of CD4 T-cells that are characterized by the cytokines they produce. These cells are mainly involved in stimulating B cells to produce antibody and are often called helper T-cells. It is also known that extracellular antigens tend to stimulate the production of Th2 cells. Thus, as used herein, "Th2 cells" is interchangeable with "helper T-cells".
[0089] The term "total cholesterol" as used herein refers to the sum of three kinds of lipids: high-density lipoprotein (HDL), low-density lipoprotein (LDL), and triglycerides. Levels of serum total cholesterol of > 200 mg/dl are levels that are an indicating risk factor for atherosclerosis and cardiovascular disease.
[0090] The term "triglycerides" as used herein are composed of fatty acid molecules and are the basic chemicals contained in fats in both animals and plants.
[0091] The term "type I osteoporosis" or "postmenopausal osteoporosis" as used herein is usually found in women after the beginning of menopause. The incidence in women is six to eight times higher than that in men. It has been postulated that the cause of this osteoporosis is accelerated bone resorption. The increased bone turnover results in a secondary decrease in parathyroid hormone (PTH) secretion as well as a secondary reduction in the renal production of calcitriol. Patients present with trabecular bone loss with vertebral fractures or distal forearm fractures.
[0092] The term "type II osteoporosis" or "age-associated osteoporosis" or "senile osteoporosis" occurs in men or women over the age of 70. The mechanisms of this bone mass loss are thought to be increased PTH secretion resulting from decreased gastrointestinal calcium absorption and decreased osteoblast function. Patients usually present with fractures of the hip or vertebrae, sites that contain cortical and trabecular bone, although fractures of the pelvis, ribs, and tibia can also occur.
[0093] The term "very low density lipoprotein" or "VLDL" as used herein refers to a triglyceride carrying lipoprotein. I. Lactoferrin
[0094] The lactoferrin used according to the present invention can be obtained through isolation and purification from natural sources, for example, but not limited to mammalian milk. The lactoferrin is preferably mammalian lactoferrin, such as bovine or human lactoferrin. In preferred embodiments, the lactoferrin is produced recombinantly using genetic engineering techniques well known and used in the art, such as recombinant expression or direct production in genetically altered animals, plants or eukaryotes, or chemical synthesis. See, i.e., U.S. Patent Nos. 5,571,896; 5,571,697 and 5,571,691, which are herein incorporated by reference.
[0095] In certain aspects, the present invention provides lactoferrin variants having enhanced biological activities of natural LF and or rLF, e.g., the ability to stimulate and/or inhibit cytokines or chemokines or the ability of lactoerrin and/or its variants to function as a chemokine. In particular, the invention provides variants of lactoferrin from which at least the N-terminal glycine residue has been substituted and/or truncated. The N-terminal lactoferrin variants may occur naturally or may be modified by the substitution or deletion of one or more amino acids.
A. Lactoferrin Related Peptides
[0096] In certain embodiments, the present invention concerns novel peptide compositions. As used herein, a "peptide," "amino acid molecule," "polypeptide", "peptide composition" generally refers, but is not limited to, a peptide of about 33 amino acids. As used herein, all the terms related "peptide" are interchangeable.
[0097] More particularly, the peptides of the present invention are derived from lactoferrin. Thus, a lactoferrin related peptide includes related-compounds of the respective molecules that exhibit at least some biological activity in common with their native counterparts, for example lactoferrin. Such related-compounds include, but are not limited to, truncated polypeptides and polypeptides having fewer amino acids than the native polypeptide. The full length lactoferrin protein, which includes polypeptide sequences, for example, but are not limited to SEQ.ID.NO.12 (GenBank accession AAA36159.1), SEQ.ID.NO.13 (GenBank accession CAA38572.1); SEQ.ID.NO.14 (GenBank accession NP_032548.2); SEQ.ID.NO.15 (GenBank accession NP_002334.1); SEQ.ID.NO.16 (GenBank accession NP_851341.1); SEQ.ID.NO.17 (GenBank accession AANl 1304.1); SEQ.ID.NO.18 (GenBank accession AAN75578.2); SEQ.ID.NO.19 (GenBank accession CAA06441.1); SEQ.ID.NO.20 (GenBank accession BAA13633.1); SEQ.ID.NO.21 (GenBank accessionAAG48753.1); SEQ.ID.NO.22 (GenBank accession BAB03470.1); SEQ.ID.NO.23 (GenBank accession AAF82241.1); SEQ.ID.NO.24 (GenBank accession CAB53387.1); SEQ.ID.NO.25 (GenBank accession BAA07458.1); SEQ.ID.NO.26 (GenBank accession CAA09407.1); SEQ.ID.NO.27 (GenBank accession AAB60324.1); SEQ.ID.NO.28 (GenBank accession CAA55517.1); SEQ.ID.NO.29 (GenBank accession CAA40366.1); SEQ.ID.NO.30 (GenBank accession CAA37914.1); SEQ.ID.NO.31 (GenBank accession CAA37116.1); SEQ.ID.NO.32 (GenBank accession AAA97958.1); SEQ.ID.NO.33 (GenBank accession AAA59479.1); SEQ.ID.NO.34 (GenBank accession AAA59511.1); SEQ.ID.NO.35 (GenBank accession AAA30610.1); SEQ.ID.NO.36 (GenBank accession AAA31102.1); SEQ.ID.NO.37 (GenBank accession AAA31059.1); SEQ.ID.NO.38 (GenBank accession AAA30617.1); and SEQ.ID.NO.39 (GenBank accession AAA30609.1), more fully described in U.S. Patent Nos. 6,399,570; 5,304,633; 5,317,084; 5,656,591; 5,849,885; 5,849,881; 5,766,939 which are incorporated herein by reference in its entirety. More specifically, the lactoferrin related peptides may include the following sequences, but are not limited to these sequences, SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP-31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO;8 (LFP-33), SEQJD.NO.9 (LFP-34), SEQ.ID.NO.IO (LFP-35), and SEQ.ID.NO.i l (LFP-36).
[0098] In certain embodiments the size of the peptide composition or amino acid molecule may comprise, but is not limited to, about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 60 or greater amino molecule residues, and any range derivable therein.
[0099] As used herein, an "amino molecule" refers to any amino acid, amino acid derivative or amino acid mimic as would be known to one of ordinary skill in the art. In certain embodiments, the residues of the peptide are sequential or contiguous, without any non-amino molecule interrupting the sequence of amino molecule residues. In other embodiments, the sequence may comprise one or more non-amino molecule moieties. In particular embodiments, the sequence of residues of the peptide may be interrupted by one or more non-amino molecule moieties.
[0100] Accordingly, the term "peptide" encompasses amino molecule sequences comprising at least one of the 20 common amino acids in naturally synthesized proteins, or at least one modified or unusual amino acid, including but not limited to those shown on Table 1 below.
Figure imgf000025_0001
[00101] Peptide compositions may be made by any technique known to those of skill in the art, including the expression of proteins, polypeptides or peptides through standard molecular biological techniques, the isolation of peptide compounds from natural sources, or the chemical synthesis of peptides. The nucleotide and protein, polypeptide and peptide sequences for various genes have been previously disclosed, and may be found at computerized databases known to those of ordinary skill in the art. One such database is the National Center for Biotechnology Information's Genbank and GenPept databases. The coding regions for these known genes may be amplified and/or expressed using the techniques disclosed herein or as would be know to those of ordinary skill in the art. Alternatively, various commercial preparations of polypeptides and peptides are known to those of skill in the art.
[0102] In certain embodiments a peptide composition may be purified. Generally, "purified" will refer to a specific polypeptide, or peptide composition that has been subjected to fractionation to remove various other proteins, polypeptides, or peptides, and which composition substantially retains its activity, as may be assessed, for example, by the protein assays, as would be known to one of ordinary skill in the art for the specific or desired polypeptide or peptide.
B. Variants
[0103] Deletional variants can be produced by proteolysis of lactoferrin and/or expression of a polynucleotide encoding a truncated lactoferrin as described in U.S. Patent 6,333,311, which is incorporated herein by reference.
[0104] Substitutional variants or replacement variants typically contain the exchange of one amino acid for another at one or more sites within the protein. Substitutions can be conservative, that is, one amino acid is replaced with one of similar shape and charge. Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine. Substitutional variants or replacement can be produced using Standard mutagenesis techniques, for example, site-directed mutagenesis as disclosed in U.S. Patents 5,220,007; 5,284,760; 5,354,670; 5,366,878; 5,389,514; 5,635,377; 5,789,166, and 6,333,311, which are incorporated herein by reference. [0105] In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
[0106] Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics (Kyte and Doolittle, 1982), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (- 1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
[0107] It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, e.g., still obtain a biological functionally equivalent protein. In making such changes, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those that are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
[0108] It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Patent 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein. As detailed in U.S. Patent 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ± 1); glutamate (+3.0 ± 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ± 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4).
[0109] Still further, it is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtains a biologically equivalent and immunologically equivalent protein. In such changes, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those that are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
[0110] Other types of variants can include variants in which at least the N-terminal glycine amino acid residue can be replaced or substituted with any of the twenty natural occurring amino acids, for example a positively charged amino acid (arginine, lysine, or histidine), a neutral amino acid (alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylaline, proline, serine, threonine, tryptophan, tyrosine, valine) and/or a negatively charged amino acid (aspartic acid or glutamic acid). Still further, it is contemplated that any amino acid residue within the range of Nl to Nl 6 can be replaced or substituted. It is envisioned that at least up to 16 of the N-terminal amino acids residues can be replaced or substituted as long as the protein retains it biological and/or functional activity, which is stimulating the production of various cytokines, such as IL-18, MIP-3α, GM-CSF or IFN-γ, or inhibiting production of various cytokines, such as IL-2, IL-4, IL-5, IL-10, and TNF-α. Thus, the N-terminal lactoferrin variants of the present invention are considered functional equivalents oflactoferrin.
[0111] Still further, the N-terminal amino acid residues can be substituted with a modified and/or unusual amino acids. A table of exemplary, but not limiting, modified and/or unusual amino acids is provided herein below.
[0112] The presence and the relative proportion of an N-terminal lactoferrin variants (deletions and/or substitutions) in a preparation of lactoferrin (lactoferrin composition) may be done by determination of the N-terminal amino acid sequence by the process of Edman degradation using standard methods. A relative proportion of N-terminal lactoferrin variant comprises at least 1% of the lactoferrin composition, at least 5% of the lactoferrin composition, at least 10% of the lactoferrin composition, at least 25% of the lactoferrin composition, at least 50% of the lactoferrin composition or any range in between.
[0113] In this method, the protein is reacted with phenylisothiocyanate (PITC), which reacts with the amino acid residue at the amino terminus under basic conditions to form a phenylthiocarbamyl derivative (PTC-protein). Trifluoroacetic acid then cleaves off the first amino acid as its anilinothialinone derivative (ATZ-amino acid) and leaves the new amino terminus for the next degradation cycle.
[0114] The percentage of N-terminal lactoferrin variant may also be done more precisely by using a Dansylation reaction. Briefly, protein is dansylated using Dansyl chloride reacted with the protein in alkaline conditions (pH 10). Following the Dansylation, the reaction mixtures are dried to pellets, then completely hydrolyzed in 6N HCl. The proportion of N- terminal amino acids are identified by RP HPLC using an in-line fluorometer in comparison with standards made up of known dansylated amino acids.
[0115] In terms of functional equivalents, it is well understood by the skilled artisan that, inherent in the definition of a "biologically functional equivalent" protein is the concept that there is a limit to the number of changes that may be made within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity and/or enhancing the biological activity of the lactoferrin molecule. Biologically functional equivalents are thus defined herein as those proteins in which selected amino acids (or codons) may be substituted. Functional activity is defined as the ability of the lactoferrin related peptide to stimulate or inhibit various cytokines or chemokines.
[0116] Yet further, the lactoferrin peptides of this invention can be in glycosylated or unglycosylated form, can be modified post-translationally (e.g., acetylation, and phosphorylation) or can be modified synthetically (e.g., the attachment of a labeling group). Fragments of lactoferrin molecules that retain the prophylactic efficacy can also be used (see, e.g. PCT/IB 00/00271, which is incorporated herein in reference in its entirety).
C. Production of Lactoferrin
1. Synthetic Variants
[0117] The present invention also describes lactoferrin, lactoferrin variants and lactoferrin related peptides for use in various embodiments of the present invention. Because of their relatively small size, the peptides of the invention can also be synthesized in solution or on a solid support in accordance with conventional techniques. Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, for example, Stewart and Young (1984); Tarn et al. (1983); Merrifield (1986); and Barany and Merrifield (1979), each incorporated herein by reference. Short peptide sequences, or libraries of overlapping peptides, usually from about 6 up to about 35 to 50 amino acids, which correspond to the selected regions described herein, can be readily synthesized and then screened in screening assays designed to identify reactive peptides. Alternatively, recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a peptide of the invention is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression.
2. Recombinant Synthesis a) Nucleic Acid Compositions
[0118] Certain embodiments of the present invention concern a nucleic acid composition having a nucleic acid sequence of lactoferrin. In certain aspects, a lactoferrin nucleic acid comprises a wild-type or a mutant lactoferrin nucleic acid. In particular aspects, a lactoferrin nucleic acid sequence encodes for or comprises a transcribed nucleic acid. In other aspects, a lactoferrin nucleic acid sequence comprises a nucleic acid segment of SEQ.ID.NO.40 (GenBank accession M93150); SEQ.ID.NO.41 (GenBank accession X54801); SEQ.ID.NO.42 (GenBank accession NM_008522); SEQ.ID.NO.43 (GenBank accession NM_002343); SEQ.ID.NO.44 (GenBank accession NM_180998); SEQ.ID.NO.45 (GenBank accession AΫ137470); SEQ.ID.NO.46 (GenBank^ accession AYl 78998); " SEQ~JD.NO.47~ (GenBank accession AJ005203); SEQ.ID.NO.48 (GenBank accession D88510); SEQ.ID.NO.49 (GenBank accession AF332168); SEQ.ID.NO.50 (GenBank accession AB046664); SEQ.ID.NO.51 (GenBank accession AAF82241.1); SEQ.ID.NO.52 (GenBank accession AJ131674); SEQ.ID.NO.53 (GenBank accession D38380); SEQ.ID.NO.54 (GenBank accession AJOl 0930); SEQ.ID.NO.55 (GenBank accession U07643); SEQ.ID.NO.56 (GenBank accession X78902); SEQ.ID.NO.57 (GenBank accession X57084); SEQ.ID.NO.58 (GenBank accession X53961); SEQ.ID.NO.59 (GenBank accession X52941); SEQ.ID.NO.60 (GenBank accession U53857); SEQ.ID.NO.61 (GenBank accession M73700); SEQ.ID.NO.62 (GenBank accession M83202); SEQ.ID.NO.63 (GenBank accession L19981); SEQ.ID.NO.64 (GenBank accession M92089); SEQ.ID.NO.65 (GenBank accession M81327); SEQ.ID.NO.66 (GenBank accession M63502); and SEQ. ID .NO.67 (GenBank accession L08604) or a biologically functional equivalent thereof. In particular aspects, the nucleic acid sequence of the present invention encodes a polypeptide, peptide, for example, but not limited to SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP- 31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO.8 (LFP-33), SEQ.ID.NO.9 (LFP-34), SEQ.ID.NO.10 (LFP-35), and SEQ.ID.NO.i l (LFP-36). Thus, nucleic acid compositions encoding lactoferrin and/or lactoferrin related peptides are herein provided and are also available to a skilled artisan at accessible databases, including the National Center for Biotechnology Information's GenBank database and/or commercially available databases, such as from Celera Genomics, Inc. (Rockville, MD).
[0119] The term "nucleic acid" is well known in the art. A "nucleic acid" as used herein will generally refer to a molecule (i.e., a strand) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase. A nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine "A," a guanine "G," a thymine "T" or a cytosine "C") or RNA (e.g., an A, a G, an uracil "U" or a C). The term "nucleic acid" encompass the terms "oligonucleotide" and "polynucleotide," each as a subgenus of the term "nucleic acid." The term "oligonucleotide" refers to a molecule of between about 3 and about 100 nucleobases in length. The term "polynucleotide" refers to at least one molecule of greater than about 100 nucleobases in length.
[0120] These definitions generally refer to a single-stranded molecule, but in specific embodiments will also encompass an additional strand that is partially, substantially or fully complementary to the single-stranded molecule. Thus, a nucleic acid may encompass a double-stranded molecule or a triple-stranded molecule that comprises one or more complementary strand(s) or "complement(s)" of a particular sequence comprising a molecule. As used herein, a single stranded nucleic acid may be denoted by the prefix "ss," a double stranded nucleic acid by the prefix "ds," and a triple stranded nucleic acid by the prefix "ts."
[0121] The nucleic acids of the present invention encompass biologically functional equivalent lactoferrin proteins, polypeptides, or lactoferrin related peptides. Such sequences may arise as a consequence of codon redundancy or functional equivalency that is known to occur naturally within nucleic acid sequences or the proteins, polypeptides or peptides thus encoded. Alternatively, functionally equivalent proteins, polypeptides or peptides may be created via the application of recombinant DNA technology, in which changes in the protein, polypeptide or peptide structure may be engineered, based on considerations of the properties of the amino acids being exchanged. Changes designed by man may be introduced, for example, through the application of site-directed mutagenesis techniques as discussed herein below, e.g., to introduce improvements or alterations to the antigenicity of the protein, polypeptide or peptide.
3. Expression Vectors
[0122] The present invention may involve using expression constructs as the pharmaceutical composition. In certain embodiments, it is contemplated that the expression construct comprises polynucleotide sequences encoding polypeptides which can enhance the immune system, for example the expression construct may comprise a nucleic acid sequence encoding the amino acid sequence of SEQ.ID.NO.l (LFP-21), SEQ.ID.NO.2 (LFP-22), SEQ.ID.NO.3 (LFP-23), SEQ.ID.NO.4 (LFP-24), SEQ.ID.NO.5 (LFP-25), SEQ.ID.NO.6 (LFP- 31), SEQ.ID.NO.7 (LFP-32), SEQ.ID.NO.8 (LFP-33), SEQ.ID.NO.9 (LFP-34), SEQ.ID.NO.10 (LFP-35), SEQ.ID.NO.i l (LFP-36), SEQ.ID.NO.12.
[0123] In certain embodiments, the present invention involves the manipulation of genetic material to produce expression constructs that encode the peptides of the present invention. Thus, the peptides are contained in an expression vector. Such methods involve the generation of expression constructs containing, for example, a heterologous nucleic acid sequence encoding the peptide of interest and a means for its expression, replicating the vector in an appropriate cell, obtaining viral particles produced therefrom, and infecting cells with the recombinant virus particles.
[0124] As used in the present invention, the term "expression vector" refers to any type of genetic construct comprising a nucleic acid coding for a lactoferrin related peptide, for example, but not limited to SEQ.ID.NO.l, SEQ.ID.NO.2, SEQ.ID.NO.3, SEQ.ID.NO.4, SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.i l. In some cases, DNA molecules are then translated into a protein, polypeptide, or peptide. Expression vectors can contain a variety of "control sequences," which refer to nucleic acid sequences necessary for the transcription and possibly translation of an operably linked coding sequence in a particular host cell. In addition to control sequences that govern transcription and translation, vectors and expression vectors may contain nucleic acid sequences that serve other functions as well and are described infra. It is contemplated in the present invention, that virtually any type of vector may be employed in any known or later discovered method to deliver nucleic acids encoding a lactoferrin and/or lactoferrin related peptide. An expression vector comprising a nucleic acid encoding a lactoferrin and/or lactoferrin related peptide may comprise a virus or engineered construct derived from a viral genome.
[0125] In particular embodiments of the invention, a plasmid vector is contemplated for use to transform a host cell. In general, plasmid vectors containing replicon and control sequences that are derived from species compatible with the host cell are used in connection with these hosts. The vector ordinarily carries a replication site, as well as marking sequences that are capable of providing phenotypic selection in transformed cells. Plasmid vectors are well known and are commercially available. Such vectors include, but are not limited to, the commercially available pSupervector (OligoEngine, Seattle, WA), and pSuppressorNeo vector (IMGENEX Corporation). Other vectors that may be employed in the present invention include, but are not limited to, the following eukaryotic vectors: pWLNEO, pSV2CAT, pOG44, PXTl, pSG (Stratagene) pSVK3, pBSK, pBR322, pUC vectors, vectors that contain markers that can be selected in mammalian cells, such as pCDNA3.1, episomally replicating vectors, such as the pREP series of vectors, pBPV, pMSG, pSVL (Pharmacia), adenovirus vector (AAV; pCWRSV, Chatterjee et al. (1992)); retroviral vectors, such as the pBABE vector series, a retroviral vector derived from MoMuLV (pGINa, Zhou et al., (1994)); and pTZ18U (BioRad, Hercules, CA).
[0126] In one embodiment, a gene encoding a lactoferrin and/or lactoferrin related peptide is introduced in vivo in a viral vector. The ability of certain viruses to enter cells via receptor-mediated endocytosis and to integrate into the host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells (Ridgeway, 1988; Nicolas and Rubenstein, 1988; Baichwal and Sugden, 1986; Temin, 1986). Such vectors include an attenuated or defective DNA virus, such as but not limited to herpes simplex virus (HSV), papilloma virus, Epstein Barr virus (EBV), adenovirus, adeno-associated virus (AAV), lentivirus and the like. Defective viruses, which entirely or almost entirely lack viral genes, are preferred. Defective virus is not infective after introduction into a cell. Use of defective viral vectors allows for administration to cells in a specific, localized area, without concern that the vector can infect other cells. Thus, any tissue can be specifically targeted. Examples of particular vectors include, but are not limited to, a defective herpes virus 1 (HSVl) vector (Kaplitt et al., 1991) an attenuated adenovirus vector, (Stratford- Perricaudet et al., 1992), a defective adeno-associated virus vector (Samulski et al., 1987 and Samulski et al., 1989) or pox- vector. Such vectors may be used to (i) transform cell lines in vitro for the purpose of expressing the a lactoferrin and/or lactoferrin related peptide or (ii) to transform cells in vitro or in vivo to provide therapeutic molecules for gene therapy. Thus, the present invention contemplates viral vectors such as, but not limited to, an adenoviral vector, an adeno-associated viral vector, a retroviral vector, a lentiviral vector, a herpes viral vector, polyoma viral vector, pox- vector or hepatitis B viral vector.
[0127] Preferably, for in vivo administration, an appropriate immunosuppressive treatment is employed in conjunction with the viral vector, e.g., adenovirus vector, to avoid immunodeactivation of the viral vector and transfected cells. For example, immunosuppressive cytokines, such as interleukin-12 (IL- 12), interferon-γ (IFN-γ), or anti-CD4 antibody, can be administered to block humoral or cellular immune responses to the viral vectors (Wilson, Nature Medicine (1995). In addition, it is advantageous to employ a viral vector that is engineered to express a minimal number of antigens.
[0128] In another embodiment the gene can be introduced in a retroviral vector, e.g., as described in U.S. Pat. No. 5,399,346; Mann et al., 1983; U.S. Pat. No. 4,650,764; U.S. Pat. No. 4,980,289; Markowitz et al., 1988; U.S. Pat. No. 5,124,263; International Patent Publication No7 WO 95707358; and Kuo et al., 1993, each of which is incorporated herein by reference in its entirety. Targeted gene delivery is described in International Patent Publication WO 95/28494.
[0129] Alternatively, the vector can be introduced in vivo by lipofection. For the past decade, there has been increasing use of liposomes for encapsulation and transfection of nucleic acids in vitro. Synthetic cationic lipids designed to limit the difficulties and dangers encountered with liposome mediated transfection can be used to prepare liposomes for in vivo transfection of a gene encoding a marker. The use of cationic lipids may promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes. The use of lipofection to introduce exogenous genes into the specific organs in vivo has certain practical advantages. Molecular targeting of liposomes to specific cells represents one area of benefit. Lipids may be chemically coupled to other molecules for the purpose of targeting. Targeted peptides, e.g., hormones or neurotransmitters, and proteins such as antibodies, or non-peptide molecules could be coupled to liposomes chemically. [0130] It is also possible to introduce the vector in vivo as a naked DNA plasmid. Naked DNA vectors for gene therapy can be introduced into the desired host cells by methods known in the art, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, use of a gene gun, or use of a DNA vector transporter (Wu and Wu, 1988).
[0131] As used herein, the terms "cell," "cell line," and "cell culture" may be used interchangeably. All of these terms also include their progeny, which is any and all subsequent generation formed by cell division. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations. A host cell may be "transfected" or "transformed," which refers to a process by which exogenous nucleic acid is transferred or introduced into the host cell. A transformed cell includes the primary subject cell and its progeny. As used herein, the terms "engineered" and "recombinant" cells or host cells are intended to refer to a cell into which an exogenous nucleic acid sequence, such as, for example, a lactoferrin and/or lactoferrin related peptide. Therefore, recombinant cells are distinguishable from naturally occurring cells that do not contain a recombinantly introduced nucleic acid.
[0132] Host cells may be derived from prokaryotes or eukaryotes, depending upon whether the desired result is replication of the vector or expression of part or all of the vector encoded nucleic acid sequences. Numerous cell lines and cultures are available for use as a host cell, and they can be obtained through the American Type Culture Collection (ATCC), which is an organization that serves as an archive for living cultures and genetic materials. In certain embodiments, a cell may comprise, but is not limited to, at least one skin, bone, neuron, axon, cartilage, blood vessel, cornea, muscle, facia, brain, prostate, breast, endometrium, lung, pancreas, small intestine, blood, liver, testes, ovaries, cervix, colon, skin, stomach, esophagus, spleen, lymph node, bone marrow, kidney, peripheral blood, embryonic or ascite cell, and all cancers thereof. An appropriate host can be determined by one of skill in the art based on the vector backbone and the desired result. Bacterial cells used as host cells for vector replication and/or expression include DH5α, JMl 09, and KC8, as well as a number of commercially available bacterial hosts such as SURE® Competent Cells and SOLOP ACK™ Gold Cells (STRATAGENE®, La JoUa). [0133] Examples of eukaryotic host cells for replication and/or expression of a vector include HeLa, NIH3T3, Jurkat, 293, Cos, CHO, Saos, and PC12. Other eukaryotic cells include yeast cells, such as Aspergillus species and Saccharomyces species. Many host cells from various cell types and organisms are available and would be known to one of skill in the art. In addition, a host cell strain may be chosen that modulates the expression of the inserted sequences, and/or modifies and/or processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and/or processing (e.g., cleavage) of protein products may be important for the function of the protein.
II. Pharmaceutical Compositions
[0134] The present invention is drawn to a composition comprising a lactoferrin composition that is dispersed in a pharmaceutical carrier. The lactoferrin that is contained in the composition of the present invention comprises lactoferrin or an N-terminal lactoferrin variant in which at least the N-I terminal glycine residue is truncated or substituted. More specifically, the N-terminal lactoferrin variant comprises at least 1% of the composition, at least 5% of the composition, at least 10% of the composition, at least 25% of the composition, at least 50% of the composition or any range in between. Yet further, the lactoferrin that is contained in the composition of the present invention comprises lactoferrin related peptides, for example, SEQ.ID.NOr 1, SEQ.ID.NO.2, SEQ.ID.NO.3Γ SEQTD.NΘ. V SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ID.NO.10, and SEQ.ID.NO.i l and/or expression vectors encoding a lactoferrin related peptide.
[0135] Yet further, the composition comprises lactoferrin in combination with a metal chelator dispersed in a pharmaceutical carrier. Thus, the present invention is drawn to a lactoferrin composition with or without a metal chelator that is dispersed in a pharmaceutical carrier. One of skill in the art understands that both compositions (e.g., lactoferrin alone or lactoferrin in combination with a metal chelator) are within the scope of the present invention and can be used interchangeably depending upon the type of response that is desired. It is envisioned that the addition of a metal chelator to the lactoferrin composition enhances the sequestering of metal ions and thus strengthens the immune system or enhances the effect of lactoferrin. [0136] Metal chelators that can be used in combination with lactoferrin, include the divalent metal chelators, for example, ethylenediaminetetraacetic acid (EDTA), [ethylenebis(oxyethylenenitrilo)] tetraacetic acid (EGTA), l,2-bis(2-aminophenoxy)ethane- N,N,N',N'-tetraacetic acid (BAPTA), hydroxyethlene triamine diacetic acid, (HEDTA) or any salts thereof. More preferably, EDTA is used in combination with lactoferrin.
[0137] In further embodiments, the present invention may concern the use of a pharmaceutical lipid vehicle compositions that include lactoferrin or lactoferrin related peptides, one or more lipids, and an aqueous solvent. As used herein, the term "lipid" will be defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds are well known to those of skill in the art, and as the term "lipid" is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man). However, a lipid is usually a biological substance. Biological lipids are well known in the art, and include for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof. Of course, compounds other than those specifically described herein that are understood by one of skill in the art as lipids are also encompassed by the compositions and methods of the present invention.
[0138] One of ordinary skill in the art would be familiar with the range of techniques that can be employed for dispersing a composition in a lipid vehicle. For example, the lactoferrin or lactoferrin related peptides may be dispersed in a solution containing a lipid, dissolved with a lipid, emulsified with a lipid, mixed with a lipid, combined with a lipid, covalently bonded to a lipid, contained as a suspension in a lipid, contained or complexed with a micelle or liposome, or otherwise associated with a lipid or lipid structure by any means known to those of ordinary skill in the art. The dispersion may or may not result in the formation of liposomes.
[0139] The composition for oral administration which is combined with a semisolid or solid carrier can be further formulated into hard or soft shell gelatin capsules, tablets, or pills. More preferably, gelatin capsules, tablets, or pills are enterically coated. Enteric coatings prevent denaturation of the composition in the stomach or upper bowel where the pH is acidic. See, e.g., U.S. Pat. No. 5,629,001. Upon reaching the small intestines, the basic pH therein dissolves the coating and permits the lactoferrin composition to be released and absorbed by specialized cells, e.g., epithelial enterocytes and Peyer's patch M cells.
[0140] Oral administration of the composition includes oral, buccal, enteral or intragastric administration. It is also envisioned that the composition may be used as a food additive. For example, the composition is sprinkled on food or added to a liquid prior to ingestion. In further embodiments, the oral composition is administered in conjunction with an antacid. Thus, an antacid is administered prior or substantially simultaneously with or after oral administration of the composition. The administration of an antacid just prior or immediately following the administration of the composition may help to reduce the degree of inactivation of the lactoferrin in the digestive tract. Examples of appropriate antacids include, but are not limited to, sodium bicarbonate, magnesium oxide, magnesium hydroxide, calcium carbonate, magnesium trisilicate, magnesium carbonate and alumin hydroxide gel.
[0141] In another embodiment, a powdered composition is combined with a liquid carrier such as, i.e., water or a saline solution, with or without a stabilizing agent.
[0142] In other preferred embodiments of the invention, lactoferrin or lactoferrin related peptides may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or inhalation.
[0143] Pharmaceutical compositions for topical administration may include the active compound formulated for a medicated application such as an ointment, paste, cream or powder. Ointments include all oleaginous, adsorption, emulsion and water-solubly based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only. Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin. Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram. Possible bases for compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base. Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the active ingredient and provide for a homogenous mixture. Transdermal administration of the present invention may also comprise the use of a "patch". For example, the patch may supply one or more active substances at a predetermined rate and in a continuous manner over a fixed period of time.
[0144] In certain embodiments, the pharmaceutical compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described e.g., in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in its entirety). Likewise, the delivery of drugs using intranasal niicroparticle resins (Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725, 871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts. Likewise, transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety).
[0145] The term aerosol refers to a colloidal system of finely divided solid of liquid particles dispersed in a liquefied or pressurized gas propellant. The typical aerosol of the present invention for inhalation will consist of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent. Suitable propellants include hydrocarbons and hydrocarbon ethers. Suitable containers will vary according to the pressure requirements of the propellant. Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
[0146] In further embodiments, the compositions of this invention, can be incorporated into various types of ophthalmic formulations for delivery to the eye (e. g., topically, intracamerally, or via an implant). The compositions are preferably incorporated into topical ophthalmic formulations for delivery to the eye. The compositions may be combined with ophthalmologically acceptable preservatives, surfactants, viscosity enhancers, penetration enhancers, buffers, sodium chloride, and water to form an aqueous, sterile ophthalmic suspension or solution. Ophthalmic solution formulations may be prepared by dissolving a compositions in a physiologically acceptable isotonic aqueous buffer. Further, the ophthalmic solution may include an ophthalmologically acceptable surfactant to assist in dissolving the compositions. Furthermore, the ophthalmic solution may contain an agent to increase viscosity, such as, hydroxymethylcellulose, hydroxyethylcellulose, hydroxypropylmethylcellulose, methylcellulose, polyvinylpyrrolidone, or the like, to improve the retention of the formulation in the conjunctival sac. Gelling agents can also be used, including, but not limited to, gellan and xanthan gum. In order to prepare sterile ophthalmic ointment formulations, the active ingredient is combined with a preservative in an appropriate vehicle, such as, mineral oil, liquid lanolin, or white petrolatum. Sterile ophthalmic gel formulations may be prepared by suspending the compositions in a hydrophilic base prepared from the combination of, for example, carbopol-974, or the like, according to the published formulations for analogous ophthalmic preparations; preservatives and tonicity agents can be incorporated.
[0147] Further, the composition for topical administration which is combined with a semi-solid carrier can be further formulated into a gel ointment. A preferred carrier for the formation of a gel ointment is a gel polymer. Gel polymers prevent denaturation of the composition in the open skin by serum proteases. The gel formulation of the present invention also provides a controlled delivery system for lactoferrin or its activity on a wound site. Controlled delivery refers to drug release or activity release sufficient to maintain a therapeutic level over an extended period of time, such as up to 24 hours or more, preferably in the range of 1 to 12 hours. The present gel formulation increases the contact time of the lactoferrin at the wound site and provides a sustained release dosage form necessary to achieve a significant increase in the rate of wound healing. This is an important advantage because it permits less frequent application of the formulation to the wound and thereby permits fewer disturbances to the wound and its cellular components.
[0148] The gel formulation of the present invention has the advantage of adhering to a wound and conforming to irregular body or wound contours. The gels may be applied directly to a wound site or in conjunction with a compliant porous or microporous substrate, for example in the form of a coating, to be applied to the wound site. Gels have the further advantages of having a high water content (which keeps the wound moist), the ability to absorb wound exudate, easy application to a wound and easy removal by washing. Gels have a cool feeling when applied to a wound and thus can increase patient comfort and acceptance of the formulation, especially on sensitive wounds. [0149] The aqueous gels of the present invention have different viscosities depending on the intended application of the gel. Viscosity is a measure of the resistance of a liquid to flow. It is defined as the ratio of the shearing stress to the rate of shearing. The shear stress is the resistance of the liquid to flow under the influence of an applied force, i.e., the molecular resistance within a body opposing an external force. The shear stress is defined as the ratio of the force to the area sheared. When a liquid is sheared, assuming laminar flow, the layers of the liquid move at different rates. The relative rate of motion of the layers is only one factor in the rate of shear. The other is the distance, or clearance between the shearing planes. Thus, shear rate is defined as the ratio of the velocity of the gel to the clearance. Viscosity has the dimensions of dynes/sec/cm2. These dimensions are referred to as poise. The dimensions of viscosity referred to herein, unless otherwise indicated, are in centipoise (cP) as measured using a Brookfield viscometer. All viscosity values are at room temperature, e.g., 220C- 25°C, unless otherwise indicated.
[0150] The gel forming materials of the present invention may be water-soluble polymers capable of forming a viscous aqueous solution or non-water soluble, water swellable polymers (e.g., collagen), which can also form a viscous solution. Swellable polymers are those that absorb water rather than dissolve in water. Cross-linked forms of the polymer described herein may not be water soluble but may be water-swellable. Therefore, cross-linked forms of the polymer are within the scope of the present invention. Cross-linking refers to covalently bonding polymer chains together with a bifunctional reagent such as glutaraldehyde. Also, it is understood by those skilled in the art that certain polymers may have to be used in the salt form or partially neutralized in order to be made water soluble. For example, it is preferable to use hyaluronic acid as sodium hyaluronate to provide suitable water solubility.
[0151] In the aqueous gel formulations for topical or incisional wound healing, the polymer may be selected from the group consisting of vinyl polymers, polyoxyethylene- polyoxypropylene copolymers, polysaccharides, proteins, poly(ethylene oxide), acrylamide polymers and derivatives or salts thereof. It is understood that poly(ethyleneoxide) includes polyethylene glycol. In the gel formulations for use in healing wounds in the anterior chamber of the eye, the polymers may be the same except that it is not preferred to use the polyoxyethylene- polyoxypropylene copolymers or poly(ethylene oxide). Also, for anterior chamber use, it is preferred that the polymer is biodegradable, i.e., it will break down into harmless constituents that can be drained from or metabolized in the anterior chamber. In the low viscosity, aqueous formulations for use in ophthalmic wound healing, the gel forming polymers may be the same as for topical or incisional wound healing, except that poly(ethylene oxide) is not preferred to be used.
[0152] The vinyl polymers useful in the present invention may be selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyvinyl pyrrolidone and polyvinyl alcohol. The polysaccharides useful in the present invention are selected from the group consisting of cellulose or cellulose derivatives, glycosaminoglycans, agar, pectin, alginic acid, dextran, starch, and chitosan. Starch occurs in two forms, α-amylose and amylopectin. The more water-soluble α-amylose is preferred. The glycosaminoglycans are selected from the group consisting of hyaluronic acid, chondroitin, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratan sulfate, heparin sulfate and heparin. The glycosaminoglycans are used to enhance wound healing in combination with any other gel-forming polymer. The proteins useful in the present invention are selected from the group consisting of collagen, gelatin and fibronectin. The acrylamide polymers are polyacrylamide or polymethacrylamide polymers. Biocompatible polyacrylamide polymers are preferred, hi further embodiments, carbomers are the preferred polyacrylamide polymer. Carbomers are synthetic high molecular weight polymers of acrylic acid cross linked with either alkyl esters of sucrose or pentaerythritol. Suitable commercially available grades of carbomer include Carbopol 910, Carbopol 934P, Carbopol 940, Carbopol 941, Carbopol 971P, Carbopol 974P, Carbopol 980, Carbopol 981, Carbopol 1342, Rheogic 252L, Rheogic 250H, and Hostacerin PN73.
[0153] In the gel formulation for topical or incisional wound healing, the viscosity may be within the range 1,000-12,000,000 cps at room temperature. It is preferred that the viscosity range be 50,000-2,000,000. In one embodiment of the present invention, the topical gel formulation may comprise 0.01-5% by weight polyacrylic acid having an average molecular weight of about 450,000-4,000,000. In a preferred embodiment, the polyacrylic acid is present at 0.5-1.5% by weight and has an average molecular weight of 2,000,000-4,000,000. The pH of the polyacrylic acid gel should be within the range 4.5-8 and more preferably in the range 6.5-7.5.
[0154] In another embodiment, the topical and incisional gel of the present invention may comprise 15-60% by weight of a polyoxyethylene-polyoxypropylene block copolymer having an average molecular weight of about 500-50,000. In a preferred embodiment, the block copolymer is present at 15-40% by weight and has an average molecular weight in the range 1,000-15,000. The block copolymers used in the present invention are commonly known as Pluronics. Preferred Pluronics are Pluronic F88 and F 127.
[0155] In a further embodiment, the topical or incisional gel may comprise 1 to 20% by weight of a cellulose polymer having a molecular weight of about 50,000 to 700,000. In a preferred embodiment, the cellulose polymer is present at 2-8% by weight and has an average molecular weight in the range 80,000-240,000. Preferred cellulose polymers are hydroxypropylmethyl cellulose (HPMC), carboxymethyl cellulose (CMC) and methyl cellulose (MC).
[0156] In a further embodiment, the topical and incisional gel may comprise 0.5- 10% by weight of hyaluronic acid having an average molecular weight in the range 500,000 to 8,000,000. In a preferred embodiment, the hyaluronic acid is present at 1.5-6.0% by weight and the average molecular weight is greater than 1,000,000.
[0157] Acrylamide polymers may be useful for all types of wound healing, particularly in the anterior chamber of the eye. An absorbable acrylamide polymer, such as polyacrylamide, may be a good substitute for present carrier systems used in ophthalmic applications, such as hyaluronic acid. The acrylamide polymers may have an average molecular weight in the range 1-13 million, preferably about 4-6 million. The weight percent of the acrylamide polymer in the gel may be 2-5%, preferably 3.5-4.5%. Substituted acrylamide polymers, such as methyl and alkyl substituted polymers are also within the scope of the present invention.
[0158] For use in the anterior chamber of the eye, an acrylamide gel delivery system has the following characteristics: any products of the dissolution or degradation of the delivery matrix are nontoxic and do not clog the trabecular mesh work; the gel is optically transparent; and the gel can be left in the anterior chamber without causing adverse clinical effects such as an unacceptable increase in ocular pressure.
[0159] It will be readily apparent to one skilled in the art that the desired viscosity range may be achieved by varying the molecular weight and percent concentration of the polymer in the formulation. For example, a gel having a low viscosity may be achieved by using a low molecular weight polymer or a lower percent concentration or a combination of the two. A high viscosity gel may be achieved by using a higher molecular weight polymer and a higher percent concentration. Intermediate viscosities may be achieved by varying the molecular weight and percent concentration accordingly.
[0160] The low viscosity solution may comprise 0.01-2.0% by weight polyacrylic acid having an molecular weight of about 100,000-4,000,000. In a preferred embodiment, the polymer is present at 0.05-0.5%. In another embodiment, this dilute viscous solution may comprise 2-40% by weight of a polyoxyethylene-polyoxypropylene copolymer having an average molecular weight of 500-500,000. Preferably, the concentration is 2-20% and the molecular weight is 1,000-15,000. Alternatively, the dilute viscous solution may comprise a cellulose polymer at 1-20% and having a molecular weight of about 80,000-240,000. It is preferred that the concentration be in the range of 1-10%. In a further embodiment, the dilute viscous solution may comprise 0.5-5.0% by weight hyaluronic acid having an average molecular weight of about 500,000-8,000,000. Preferably, the concentration is 0.5-2.0% and the average molecular weight is 1,000,000-6,000,000. If the dilute viscous solution is to be used as eye drops, it is preferred that the viscosity be in the range 1-100 cps. If it is used for other applications, such as soaking a bandage, then any viscosity in the range 1.0-5,000 will be suitable. [0147] Upon formulation, solutions are administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective to result in an improvement or remediation of the symptoms. The formulations are easily administered in a variety of dosage forms such as ingestible solutions, drug release capsules, gel ointments and the like. Some variation in dosage can occur depending on the condition of the subject being treated. The person responsible for administration can, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations meet sterility, general safety and purity standards as required by FDA Office of Biologies standards.
III. Treatment
[0161] In accordance with the present invention, a lactoferrin composition provided in any of the above-described pharmaceutical carriers is orally, topically, or parenterally administered to a subject suspected of or having a skin, fibrotic, wound, inflammatory, autoimmune, or hyperproliferative condition. One of skill in the art can determine the therapeutically effective amount of lactoferrin to be administered to a subject based upon several considerations, such as absorption, metabolism, method of delivery, age, weight, disease severity and response to the therapy. Oral administration of the lactoferrin composition includes oral, buccal, enteral or intragastric administration. It is also envisioned that the composition may be used as a food additive. For example, the composition is sprinkled on food or added to a liquid prior to ingestion. Topical administration of the lactoferrin composition includes topical, dermal, epidermal, or subcutaneous administration. Parenteral administration includes intramuscular, intravenous, intraperitoneal, and intraocular administration.
[0162] Still yet, a further embodiment is a method of using lactoferrin to modulate the immune system through its ability to both function directly as a chemokine and to modulate other key chemokines through inducing or inhibiting chemokine production or inhibiting chemokine activity. The other chemokines include but are not limited to MIP-I alpha, MIP- lβ, IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha, TECK, I-TAC, Lymphotactin, PARC, MIP-ldelta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO-α, GRO-β, GRO-γ, SCF, MEC/CCL28.The use of this constellation of properties to treat diseases with an immune component is envisaged. Many diseases or conditions have an inflammatory or immunological component where a complex network of cytokines, chemokines, and growth factors is present, modulating the migration and/or activation of different inflammatory or immune cells. It is contemplated that lactoferrin changes the balance of the cytokine, chemokine and factor milieu in favor of a therapeutic effect.
[0163] Although receptors to lactoferrin have not been fully characterized or identified, different tissues or cell types appear to express their own lactoferrin receptors and at least some of these may be involved in the modulation of immune, inflammatory, or growth responses [Suzuki and Lonnerdal 2002]. The receptors involved in this method include but are not limited to IL-8 alpha receptor, IL-8 beta receptor, neuropeptide Yl receptor, PACAP receptor, K+ channel receptor, CCR2, CCR3,, CCR6, CXCR4, EGFR, TLR-2, TLR-3, TLR-4, TLR4/MD, Mannose receptor, CD 14, TRAIL R4.
[0164] There is strong evidence that lactoferrin affects cell-mediated host responses. LF has been shown to enhance the delayed-type hypersensitivity (DTH) response in mice, mediated by macrophages and T cells, up-regulating IL-15, MIP-lalpha, and MIP-2 (Actor 2002). Lactoferrin has a high affinity for surface receptors found on activated, but not resting, lymphocytes in vitro [Spik 1994]. LF also accelerates T-cell maturation by inducing the expression of the CD4 surface marker in vitro [Dhennin-Duthille 2000]. LF has been shown to activate natural killer NK cells, induce colony-stimulating activity, activate polymorphonuclear cells (PMNs), regulate granulopoeisis, enhance antibody-dependent cell cytotoxicity, stimulate lymphokine-activated killer cell (LAK) activity, and potentiate macrophage toxicity [Shau 1992, Horowitz 1984, Gahr 1991]. There is a strong possibility that some of the immunomodulatory properties attributed to lactoferrin follow stimulation of IL- 18, a key ThI cytokine and a strong inducer of interferon-gamma and GM-CSF release [Okamura 1995, Ushio 1996, Biet 2002]. Innate and cell-mediated immunity are both known to be critical for protection against viruses and diseases such as cancer.
[0165] Indeed, LF can exert an anti-cancer effect. Lactoferrin has a significant effect on NK cells cytotoxicity against hematopoietic and breast epithelial cell lines [Bezault 1994]. Research by Kuhara, et al. [Kuhara 2000] first linked the oral administration of LF, induction of IL- 18, immunomodulation, and protection against experimental cancer metastasis (lung colonization by colon 26 carcinoma). LF increased CD4+, CD8+ and NK cells in peripheral blood of tumor bearing mice (P<0.01). Furthermore, the cytotoxic activity of NK cells from white blood cells (WBC) and splenic cells obtained from mice fed LF was enhanced when tested with cultured Ϋac-1 and Co26Lu carcinoma cells [Kuhara, et al. 2000]. Oral LF has been also reported to exhibit an anti-angiogenesis activity that could be involved in inhibiting tumor growth and metastasis [Norrby 2001]. Finally, LF can directly mediate cell cycle arrest of cancer cells via inhibition of Gl cyclin-dependent kinases, which in addition could render such cells more vulnerable to the triggering of apoptosis [Damiens 1999].
[0166] LF has been shown to protect against pathogens via its immunomodulatory activity. LF prolonged survival and decreased splenic viral titers in vivo in a mouse leukemia model of Friend virus complex (FVC-P) and was synergistic with IFN-gamma. LF and rmuIFN- gamma returned NK cell activity, decreased by FVC-P, to normal levels [Lu 1991]. Protection against a potentially lethal infection with murine Cytomegalovirus (CMV) was also observed in vivo and appeared to be mediated by an up-regulation of NK cells that was dependent on T cells [Shimizu 1996]. The immunomodulatory activity of LF and derived peptides could also elicit innate and adaptive immune responses against fungi and bacteria, including NK cells, polymorphonuclear cells, monocyte/macrophages, and T cells [Mann 1999, Wakabayashi 2002]. [0167] LF has several anti-inflammatory properties. Previous clinical studies with lactoferrin suggest that it appears to reduce gut inflammation in patients with severe graft-vs.- host disease (GVHD) [Inoue 2001]. Th2-dominated responses are involved in the pathogenesis of GVDH and thus a reversal towards a ThI environment appears to be protective [Singh 1999, Romagnani 2000]. LF stimulates a shift in immune responses from Th2 to ThI by up-regulating IL-18, IFN-gamma, and down-regulating IL-4, IL-5, and IL-IO [Zhang 2001, Guillen 2002]. Consequently, this switch to a ThI response might be the mechanism by which LF mediates the anti-GVDH effect. In other anti-inflammatory studies, oral LF was shown to protect piglets against bacterial lipopolysaccharide (LPS) -induced sepsis [Lee 1998]. Similar protection against LPS-induced mortality as well as inhibition of key inflammatory cytokines was observed with oral lactoferrin. Sepsis is mediated by TNF-alpha and studies with mice and human samples suggest that lactoferrin inhibits the in vivo production of TNF-alpha from local sources such as keratinocytes [Cumberbatch 2000, Kimber 2000, Kimber 2002].
[0168] In yet another anti-inflammatory area, human LF has been tested in a sheep model of asthma for its ability to inhibit antigen-induced bronchoconstriction (Elrod 1997). These sheep typically develop early- and late- phase bronchoconstriction and an associated increase in bronchial responsiveness following antigen challenge. Sheep treated with lactoferrin had substantial reductions in late-phase bronchoconstriction, airway hyperresponsiveness and inflammatory cell infiltration following antigen challenge compared to untreated sheep [unpublished data]. As in the case of GVHD, the pathogenesis of asthma appears to involve a predominance of Th2 immune responses [Elias 2001] and therefore the ability of LF to shift the immune response towards a ThI type may explain its anti-asthmatic effect.
[0169] The route of administration will vary, naturally, with the location and nature of the lesion, and include, for example parentally, orally or topically. Parenteral administrations include, but are not limited to intradermal, transdermal, intravenous, intramuscular, intranasal, subcutaneous, percutaneous, intratracheal, intraocular, intraperitoneal, intratumoral, perfusion, lavage, direct injection U.S. Pat. Nos. 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363 (each specifically incorporated herein by reference in its entirety). Alimentary administrations include, but are not limited to orally, buccally, rectally, or sublingually. W
[0170] Treatment methods will involve treating an individual with an effective amount of a lactoferrin composition as defined in the present invention. An effective amount is described, generally, as that amount sufficient to detectably and repeatedly to ameliorate, reduce, minimize or limit the extent of a disease or its symptoms.
[0171] The effective amount or "therapeutically effective amounts" of the lactoferrin composition to be used are those amounts effective to produce beneficial results, particularly with respect to cancer treatment, in the recipient animal or patient. Such amounts may be initially determined by reviewing the published literature, by conducting in vitro tests or by conducting metabolic studies in healthy experimental animals. Before use in a clinical setting, it may be beneficial to conduct confirmatory studies in an animal model, preferably a widely accepted animal model of the particular disease to be treated. Preferred animal models for use in certain embodiments are rodent models, which are preferred because they are economical to use and, particularly, because the results gained are widely accepted as predictive of clinical value.
[0172] As is well known in the art, a specific dose level of active compounds such as the lactoferrin composition for any particular patient depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy. The person responsible for administration will determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologies standards.
[0173] A therapeutically effective amount of a lactoferrin composition as a treatment varies depending upon the host treated and the particular mode of administration. In one embodiment of the invention the dose range of a lactoferrin composition will be about 0.5 mg/kg body weight to about 500 mg/kg body weight. The term "body weight" is applicable when an animal is being treated. When isolated cells are being treated, "body weight" as used herein should read to mean "total cell body weight". The term "total body weight" may be used to apply to both isolated cell and animal treatment. AU concentrations and treatment levels are expressed as "body weight" or simply "kg" in this application are also considered to cover the analogous "total cell body weight "and" total body weight" concentrations. However, those of skill will recognize the utility of a variety of dosage range, for example, O.lμg/kg body weight to 1 μg/kg body weight, 1 μg/kg to 1 mg/kg body weight, 1 mg/kg body weight to 450 mg/kg body weight, 2 mg/kg body weight to 400 mg/kg body weight, 3 mg/kg body weight to 350 mg/kg body weight, 4 mg/kg body weight to 300 mg/kg body weight, 5 mg/kg body weight to 250 mg/kg body weight, 6 mg/kg body weight to 200 mg/kg body weight, 7 mg/kg body weight to 150 mg/kg body weight, 8 mg/kg body weight to 100 mg/kg body weight, or 9 mg/kg body weight to 50 mg/kg body weight. Further, those of skill will recognize that a variety of different dosage levels will be of use, for example, 0.1 μg/kg, 1 μg/kg, lOμg/kg, lOOμg/kg, 200μg/kg; 300μg/kg; 400 μg/kg, 500 μg/kg, 600 μg/kg, 700 μg/kg, 800 μg/kg, 900 μg/kg, 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 7.5 mg/kg, 10 mg/kg, 12.5 mg/kg, 15 mg/kg, 17.5 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg, 40 mg/kg, 45 mg/kg, 50 mg/kg, 60 mg/kg, 70 mg/kg, 80 mg/kg, 90 mg/kg, 100 mg/kg, 120 mg/kg, 140 mg/kg, 150 mg/kg, 160 mg/kg, 180 mg/kg, 200 mg/kg, 225 mg/kg, 250mg/kg, 275mg/kg, 300 mg/kg, 325 mg/kg, 350 mg/kg, 375 mg/kg, 400 mg/kg, 450 mg/kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 700 mg/kg, 750 mg/kg, 800 mg/kg, 900 mg/kg, 1000 mg/kg, 1250 mg/kg, 1500 mg/kg, 1750 mg/kg, 2000 mg/kg, 2500 mg/kg, and/or 3000 mg/kg. Of course, all of these dosages are exemplary, and any dosage in-between these points is also expected to be of use in the invention. Any of the above dosage ranges or dosage levels may be employed for the lactoferrin composition of the present invention.
[0174] The treatments may include various "unit doses." Unit dose is defined as containing a predetermined quantity of the therapeutic composition calculated to produce the desired responses in association with its administration, i.e., the appropriate route and treatment regimen. The quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts. Also of import is the subject to be treated, in particular, the state of the subject and the protection desired. A unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time.
[0175] In certain embodiments, the composition is given in a single dose or multiple doses. The single dose may be administered daily, or multiple times a day, or multiple times a week, or monthly or multiple times a month. In a further embodiment, the composition is given in a series of doses. The series of doses may be administered daily, or multiple times a day, weekly, or multiple times a week, or monthly, or multiple times a month. A. Hyperproliferative Disease
[0176] The hyperproliferative disease, includes but is not limited to neoplasms. A neoplasm is an abnormal tissue growth, generally forming a distinct mass that grows by cellular proliferation more rapidly than normal tissue growth. Neoplasms show partial or total lack of structural organization and functional coordination with normal tissue. These can be broadly classified into three major types. Malignant neoplasms arising from epithelial structures are called carcinomas, malignant neoplasms that originate from connective tissues such as muscle, cartilage, fat or bone are called sarcomas and malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) including components of the immune system, are called leukemias, lymphomas and myelomas. A tumor is the neoplastic growth of the disease cancer. As used herein, a "neoplasm", also referred to as a "tumor", is intended to encompass hematopoietic neoplasms as well as solid neoplasms. Examples of neoplasms include, but are not limited to melanoma, non-small cell lung, small-cell lung, lung, hepatocarcinoma, retinoblastoma, astrocytoma, gliobastoma, gum, tongue, leukemia, neuroblastoma, head, neck, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, sarcoma, cervical, gastrointestinal, lymphoma, brain, colon, bladder, myeloma, or other malignant or benign neoplasms.
[0177] Other hyperproliferative diseases include, but are not limited to neurofibromatosis, rheumatoid arthritis, Waginer's granulomatosis, Kawasaki's disease, lupus erathematosis, midline granuloma, inflammatory bowel disease, osteoarthritis, leiomyomas, adenomas, lipomas, hemangiomas, fibromas, vascular occlusion, restenosis, atherosclerosis, preneoplastic lesions, carcinoma in situ, oral hairy leukoplakia, or psoriasis, and pre-leukemias, anemia with excess blasts, and myelodysplastic syndrome.
[0178] Particular neoplasms of interest in the present invention include, but are not limited to hematopoietic neoplasms. For example, a hematopoietic neoplasm may include acute myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, chronic lymphocytic leukemia or other malignancy of hematologic origin. [0179] In a preferred embodiment of the present invention, the lactoferrin compositions are administered in an effective amount to decrease, reduce, inhibit or abrogate the growth of a tumor. The amount may vary from about 0.1 μg to about lOOg of the lactoferrin composition. Preferably, the lactoferrin composition is orally administered in the range of lmg to lOOg per day, more preferably about 20mg to about 1Og per day with the most preferred dose being 4.5g per day. Intravenously administered lactoferrin can be in the range of 0.1 μg to about to 1Og per day, more preferably about 0.1 μg to about lmg with the most preferred dose being 250 mg per day. Preferably, a lactoferrin composition is intratumorally administered in the range of 0.1 μg to 1Og per day with the most preferred dose being lOOμg per day. Topically, the amount of the lactoferrin composition may vary from about lμg to about 10Og. Preferably, the topical gel, solution, capsule or tablet comprises a concentration of about 0.01% to about 20% of lactoferrin related peptides and/or lactoferrin N-terminal variants. More preferably, the topical gel, solution, capsule or tablet may comprise a concentration of about 1% to about 8.5% lactoferrin related peptides and/or lactoferrin N-terminal variants.
[0180] In certain embodiments intratumoral administration of the composition includes intratumoral injection, electroporation, or surgical or endoscopic implantation. Intratumoral injection, or injection into the tumor vasculature is specifically contemplated for discrete, solid, accessible lumorsT Local, regional or systemic administration also may be appropriate.
[0181] The tumor being treated may not, at least initially, be resectable. Treatments with the lactoferrin composition may increase the resectability of the tumor due to shrinkage at the margins or by elimination of certain particularly invasive portions. Following treatments, resection may be possible. Additional treatments subsequent to resection will serve to eliminate microscopic residual disease at the tumor site.
[0182] Alternatively, the present invention may be used at the time of surgery, and/or thereafter, to treat residual or metastatic disease. For example, a resected tumor bed may be injected or perfused with a formulation comprising the lactoferrin composition. The perfusion may be continued post-resection, for example, by leaving a catheter implanted at the site of the surgery. Periodic post-surgical treatment is also envisioned. [0183] Continuous administration also may be applied where appropriate, for example, where a tumor is excised and the tumor bed is treated to eliminate residual, microscopic disease. Delivery via syringe or catherization is preferred. Such continuous perfusion may take place for a period from about 1-2 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 weeks or longer following the initiation of treatment. Generally, the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs. It was further contemplated that limb perfusion may be used to administer therapeutic compositions of the present invention, particularly in the treatment of melanomas and sarcomas.
[0184] Still yet, a further embodiment is a method of enhancing a mucosal immune response in the gastrointestinal tract in a subject comprising the step of administering orally to said subject a lactoferrin composition. It is also envisioned that the lactoferrin composition stimulates interleukin-18 following oral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
[0185] A further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing the systemic immune system "by increasing the amount of the lactoferrin composition in the systemic circulation. Preferably, the composition is administered intravenously. It is envisioned that the lactoferrin composition stimulate interleukin-18 following intravenous administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
[0186] A further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing a local or systemic immune system by increasing the amount of lactoferrin in the vicinity of the tumor. Vicinity of the tumor refers to the general area of the tumor, for example the lactoferrin can be administered directly into or on the tumor, or in the general area of the tumor, but not directly into the tumor. The general area may include the margin area or near or adjacent the margin area of the tumor. Preferably, the lactoferrin composition is administered intratumorally. It is envisioned that the lactoferrin composition stimulates interleukin-18 following intratumoral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
[0187] A further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing a local or systemic immune system by increasing the amount of a lactoferrin composition in the skin in the vicinity of the tumor. Preferably, the composition is administered topically. As above, administration in the vicinity of the tumor includes administration near or adjacent to the margins of the tumor or directly in the margin area of the tumor. It is envisioned that the lactoferrin composition stimulates interleukin-18 and GM-CSF in the local tissue (e.g., keratinocytes), which enhances immune cells. It is envisioned that the lactoferrin composition stimulate interleukin-18 following intratumoral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
[0188] In certain embodiments, it may be desirable to combine the lactoferrin composition of the present invention with other agents effective in the treatment of hyperproliferative disease, such as anti-cancer agents, or with surgery. An "anti-cancer" agent is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, or increasing the lifespan of a subject with cancer. Anti-cancer agents include biological agents (biotherapy), chemotherapy agents, and radiotherapy agents. More generally, these other compositions would be provided in a combined amount effective to kill or inhibit proliferation of the cell. This process may involve administering the lactoferrin composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s). [0189] Alternatively, the lactoferrin composition of the present invention may precede or follow the other anti-cancer agent treatment by intervals ranging from minutes to weeks. In embodiments where the other anti-cancer agent and lactoferrin composition are administered or applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and lactoferrin composition would still be able to exert an advantageously combined effect on the cell. In such instances, it is contemplated that one may contact the cell with/administer both modalities within about 1-14 days of each other and, more preferably, within about 12-24 hours of each other. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations.
a) Chemotherapy
[0190] Cancer therapies also include a variety of chemical based treatments. Some examples of chemotherapeutic agents include without limitation antibiotic chemotherapeutics such as Doxorubicin, Daunorubicin, Adriamycin, Mitomycin (also known as mutamycin and/or mitomycin-C), Actinomycin D (Dactinomycin), Bleomycin, Plicomycin, plant alkaloids such as Taxol, Vincristine, Vinblastine, miscellaneous agents such as platinum based agents (e.g., Cisplatin (CDDP)), etoposide (VP 16), Tumor Necrosis Factor, and alkylating agents such as, Carmustine, Melphalan (also known as alkeran, L-phenylalanine mustard, phenylalanine mustard, L-PAM, or L-sarcolysin, (a phenylalanine derivative of nitrogen mustard), Cyclophosphamide, Chlorambucil, Busulfan (also known as myleran), taxane based agents (e.g., docetaxel) and Lomustine.
[0191] Some examples of other agents include, but are not limited to, Carboplatin, Procarbazine, Mechlorethamine, Irinotecan, Topotecan, Ifosfamide, Nitrosurea, Etoposide (VP 16), Tamoxifen, Raloxifene, Toremifene, Idoxifene, Droloxifene, TAT-59, Zindoxifene, Trioxifene, ICI 182,780, EM-800, Estrogen Receptor Binding Agents, Gemcitabinen, Navelbine, Farnesyl-protein transferase inhibitors, Transplatinum, 5-Fluorouracil, hydrogen peroxide, and Methotrexate, Temazolomide (an aqueous form of DTIC), Mylotarg, Dolastatin-10, Bryostatin, or any analog or derivative variant of the foregoing. b) Radiotherapeutic Agents
[0192] Radiotherapeutic agents and factors include radiation and waves that induce DNA damage for example, γ-irradiation, X-rays, UV-irradiation, microwaves, electronic emissions, radioisotopes, and the like. Therapy may be achieved by irradiating the localized tumor site with the above described forms of radiations. It is most likely that all of these factors effect a broad range of damage to DNA5 the precursors of DNA, the replication and repair of DNA, and the assembly and maintenance of chromosomes.
[0193] Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 weeks), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
c) Surgery
[0194] Approximately 60% of persons with cancer will undergo surgery of some type, which includes preventative, diagnostic or staging, curative and palliative surgery. Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene therapy, immunotherapy and/or alternative therapies.
[0195] Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed. Tumor resection refers to physical removal of at least part of a tumor. In addition to tumor resection, treatment by surgery includes laser surgery, cryosurgery, electrosurgery, and miscopically controlled surgery (Mohs' surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue.
[0196] Upon excision of part of all of cancerous cells, tissue, or tumor, a cavity may be formed in the body. Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy. Such treatment may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. These treatments may be of varying dosages as well. d) Other Biotherapy Agents
[0197] It is contemplated that other biological agents may be used in combination with the present invention to improve the therapeutic efficacy of treatment. These additional agents include, without limitation, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adhesion, agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers, or other biological agents, as well as biotherapy such as for example, hyperthermia.
[0198] Hyperthermia is a procedure in which a patient's tissue is exposed to high temperatures (up to 106°F). External or internal heating devices may be involved in the application of local, regional, or whole-body hyperthermia. Local hyperthermia involves the application of heat to a small area, such as a tumor. Heat may be generated externally with high- frequency waves targeting a tumor from a device outside the body. Internal heat may involve a sterile probe, including thin, heated wires or hollow tubes filled with warm water, implanted microwave antennae, or radiofrequency electrodes.
[0199] A patient's organ or a limb is heated for regional therapy, which is accomplished using devices that produce high energy, such as magnets. Alternatively, some of the patient's blood may be removed and heated before being perfused into an area that will be internally heated. Whole-body heating may also be implemented in cases where cancer has spread throughout the body. Warm-water blankets, hot wax, inductive coils, and thermal chambers may be used for this purpose.
[0200] Hormonal therapy may also be used in conjunction with the present invention. The use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or estrogen and this often reduces the risk of metastases.
[0201] Adjuvant therapy may also be used in conjunction with the present invention. The use of adjuvants or immunomodulatory agents include, but are not limited to tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-I, MlP-lbeta, MCP-I, RANTES, and other chemokines. e) Immunotherapy
[0202] Immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells. The immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell. The antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing. The antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent. Alternatively, the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target. Various effector cells include cytotoxic T cells and NK cells.
[0203] It is contemplated that vaccines that are used to treat cancer may be used in combination with the present invention to improve the therapeutic efficacy of the treatment. Such vaccines include peptide vaccines or dendritic cell vaccines. Peptide vaccines may include any tumor-specific antigen that is recognized by cytolytic T lymphocytes. Yet further, one skilled in the art realizes that dendritic cell vaccination comprises dendritic cells that are pulsed with a peptide or antigen and the pulsed dendritic cells are administered to the patient.
[0204] Examples of tumor-specific antigens that are being used as vaccines in melanoma include, but are not limited to gplOO or MAGE-3. These antigens are -being - administered as peptide vaccines and/or as dendritic cell vaccines.
B. Wounds and other Integument Disorders 1. Wounds
[0205] The present invention is designed for the treatment of any type of skin disease including but not limited to psoriasis, scleroderma, atopic dermatitis, epidermolysis bullosa, acute wounds, chronic wounds, and burns.
[0206] The present invention is designed for the treatment of any type of wound, which includes, but is not limited to skin wound, internal wound, gastrointestinal wound, oral wound, bone wounds, ophthalmic wound, surgical wound, or any combination thereof. Wounds can be found on but not limited to skin, internal organs, stomach and intestines (gastrointestinal), oral mucosa, and eye (ophthalmic wounds, e.g., corneal ulcers, radiokeratotomy, corneal transplants, epikeratophakia and other surgically induced wounds in the eye). Depending on the process that causes the wounds, wounds can also be classified as but are not limited to incisional wounds, excisional wounds, diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers, chemical wounds, burn wounds, full-thickness skin wound and partial-thickness skin wound.
[0207] A further embodiment of the present invention is administering the inventive composition to treat skin wounds. Skin wounds further comprise but are not limited to full-thickness wounds and partial-thickness wounds. Full-thickness wounds involve the complete removal of epidermis and dermis to the depth of fascial planes or subcutaneous fat. In the loose-skinned species, the thin musculature of the panniculus carnosus, which firmly adheres to the base of the dermis, is usually removed as well. In partial-thickness wounds a substantial amount of dermis, mostly reticular, is left behind, and, more importantly, the bases of most epidermal appendages (sebaceous and sweat glands, hair follicles) remain intact.
[0208] Yet further, a wound can be further defined as an acute wound. Acute wounds have a relatively rapid rate of healing, especially in healthy subjects. However, in the elderly or immunocompromised healing can be prolonged. Healing is also prolonged if the wound becomes infected. Preferred acute wounds that are to be treated with the present composition include, but are not limited to partial-thickness burns, lacerations, bullet wounds or infected wounds.
[0209] A wound is also further defined as a chronic wound. Examples of chronic wounds or chronic ulcers include, but are not limited to diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers. Yet further, chronic wounds can also include infected wounds. Chronic wounds are wounds that do not repair or do so extremely slowly, and show partial or total lack of structural organization and functional coordination with normal tissue. Chronic wounds or chronic ulcers can be broadly classified into three major types: diabetic ulcers, venous stasis ulcers, decubitus or pressure ulcers. Diabetic ulcers often occur on a foot. Chronic diabetic state and poor glucose control results in poor peripheral circulation and microcirculation due to progressive arteriosclerosis; neuropathic changes that result in an insensate extremity prone to trauma; and intrinsic defects in the wound healing process that may include reduced abundance and response to cellular growth factors. In the case of venous ulcers, venous hypertension causes disturbed microcirculation and pathological changes of the capillaries, elevated persistent levels of pro-inflammatory cytokines and proteases. Fibroblast senesce and respond less to growth factors, which distribute unfavorably. Proteolytic enzymes and their inhibitors are imbalanced. Pressure ulcers occur when skin is under pressure without movement to allow blood flow for 8-12 hours.
[0210] In a preferred embodiment of the present invention, the inventive composition is administered in an effective amount to seal, to close, to improve or to repair the wound. Also, it is envisioned that the composition of the present invention can also decrease, reduce, or inhibit, bacterial infections of the wound, which aid in the healing process of a wound. Thus, one of skill in the art realizes that depending upon the wound type, location, health of the subject, etc., the lactoferrin composition of the present invention may be administered for any given period of time until the wound is healed at least by 5%, 10%, 20%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% or any range in between.
[0211] For topical administration, the gel formulation of the present invention may be used to coat fibers of an absorbent gauze dressing to form a wound healing bandage which may then be placed on a wound. The low viscosity formulation is preferred for this use. The wound healing bandage may be prepared by soaking a gauze dressing with an aqueous gel solution containing lactoferrin having wound healing activity. The bandage can then be applied to the wound scTthat the coated fibers of the gauze contacts the wound and stimulate the rate of wound healing.
[0212] In those applications where the present invention is a gel that is applied to an internal or incisional wound, it is preferred that the gel forming polymer be biodegradable. The naturally occurring polymers are generally biodegradable. Examples of these are collagen, the glycosaminoglycans, gelatin and starch. The cellulosics are not biodegradable. The synthetic polymers such as the vinyl polymers are not degradable. The biodegradability of the polymers described herein is well known to those skilled in the art.
[0213] A further embodiment of the present invention is a method of treating a wound comprising the step of supplementing the local immune system by increasing the amount of a lactoferrin composition in the vicinity of the wound. Preferably, the peptide is administered topically to the wound. [0214] Yet further, the present invention also provides a method of treating a wound comprising the step of supplementing the systemic immune system by increasing the amount of a lactoferrin composition in the systemic circulation. Preferably, the lactoferrin is administered via a parenteral route, which includes, but is not limited to intramuscular, intravenous, intraperitoneal, intraoccular, intraarticular or into a surgical field.
[0215] In further embodiments, the present invention provides a method of enhancing the immune system of a subject suffering from a wound by administering to the subject a lactoferrin composition. Depending upon the mode of administration, different arms of the immune system are enhanced. For example, topical administration of the composition results in enhancement of the local immune system, i.e., in the vicinity of the wound. Parenteral administration of the composition results in enhancement of the systemic immune system. Yet further, oral administration of the composition results in enhancement of the mucosal immune system, which can also result in systemic effects as well. It is further contemplated that IL- 18 or GM-CSF stimulate the production or activity of cells involved in wound repair, for example, but not limited to keratinocytes, endothelial cells, dendritic cells, fibroblasts, and myofibroblasts. Yet further, it is envisioned that the lactoferrin composition inhibit the production of TNF-alpha, which may lead to excess inflammation and tissue destruction for example by stimulating the production of metalloproteinases.
[0216] It is further envisioned that supplementing the local immune system in a subject by administering topically a therapeutically effective amount of the inventive composition in the vicinity of the wound can result in the killing of bacteria infecting the wound. Still further, topical administration of a composition comprising a lactoferrin peptide related compound may stimulate the production of a cytokine or a chemokine. Cytokines, for example, interleukin-18 or granulocyte/macrophage colony-stimulating factor, can also stimulate the production or activity of cells involved in wound repair. The cells involved in wound repair include, but are not limited to keratinocytes, endothelial cells, fibroblasts, dendritic cells, and myofibroblasts. The inhibition of TNF-alpha further inhibits the migration and maturation of dendritic cells and the production of metalloproteinases. The dendritic cells can be Langerhans cells. [0217] In order to increase the effectiveness of the composition of the present invention, it may be desirable to combine the composition of the present invention with other agents effective in the treatment of wounds, such as growth factors, skin replacement therapy, enzymatic and surgical debridement, moist wound dressings, cleansers, antibiotics. Such wound healing agents are capable of negatively affecting a wound in a subject, for example, by enhancing the growth rate of skin cells, augmenting the blood supply to skin cells, promoting an immune response against bacteria infecting the wound, killing bacteria, cleaning ischemic tissue, promoting the closure of the wound. More generally, these other wound healing agents are provided in a combined amount effective to promote the healing of a wound. This process may involve administering the composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s).
[0218] Alternatively, the composition of the present invention may precede or follow the other wound healing agent treatment by intervals ranging from minutes to weeks. In embodiments where the other wound healing agent and inventive composition are administered or applied separately to the wound, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and human lactoferrin composition would still be able to exert an advantageously combined effect on the wound. In such instances, it is contemplated that one may contact the wound with/administer both modalities within about 1-14 days of each other and, more preferably, within about 12-24 hours of each other. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations.
a) Growth Factors
[0219] Wound healing therapies include growth factor based treatments. Examples include, but are not limited to Regranex™ (Becaplermin-BB gel), AuToIo-GeI (autologous activated platelet releasate), Procuren (autologous thrombin-induced platelet releasate). Growth factors act without limitation by promoting granulation or the formation of new highly vascularized connective tissue; stimulating proliferation, differentiation and migration of epithelial cells, vascular endothelial cells and other skin cells; enhancing the production of collagen, collagenase, and extracellular matrix.
b) Skin Replacement Therapy
[0220] Examples include but are not limited to Apligraf (bilayered living skin), Trancyte (Human fibroblast-derived temporary skin substitute), Dermagraft (permanent, one- layer skin substitute), Epicel (living one-layer artificial skin), Integra (collagen-based skin regeneration template), AlloDerm (single-layer artificial skin made from human cadavers), CCS (living, cultured, artificial skin).
c) Enzymatic and Surgical Debridement
[0221] Debridement is a process or procedure to clean ischemic or dead tissue. Enzymatic debriders include Accuzyme papain-urea debriding ointment and Collagenase Santyl. Surgical debridement refers to physical removal of at least part of the ischemic or dead tissue in a wound. Debridement may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. Enzymatic debridement treatments may be of varying dosages as well. It is further contemplated that the present invention may be used in conjunction with enzymatic or surgical debridement.
d) Dressings
[0222] Wound healing therapies include a variety of treatments based on dressings. Dressing categories include but are not limited to amorphous hydrogels, hydrogel sheets, absorptives, alginates, biological and synthetic dressings, collagens, composites, contact layers, elastic gauzes, foams, gauzes and non-woven dressings, hydrocolloids, impregnated dressings, silicone gel sheets, silver dressings, transparent films, wound fillers
e) Cleansers
[0223] Examples include but are not limited to Biolex, Lamin, Wound Wash Saline, Techni-Care, CarraKlenz, DiaB Klenz, MicroKlenz, RadiaCare Klenz, UltraKlenz, Comfee Sea-Clens, Optipore Sponge, Saf-Clens, Shur-Clens, Dermagran, DermaKlenz, Dumex, Gene Klenz, GRX, Allclenz, Restore, Hyperion, Medi Tech, Skintegrity, MPM Antimicrobial, Clins Wound, Septicare, Lobana Saline. f) Antimicrobials
[0224] Examples of include but are not limited to Sulfamylon Cream, Thermazene Cream (1% silver sulfadiazine), cadexomer-iodine pads or gel. Examples of intravenous antimicrobials include but are not limited to imipenem/cilastatin, β-lactam/ β-lactamase inhibitors (ampicillin/sulbactam, piperacillin/tazobactam), and broad-spectrum cephalosporins (cefoxitin, ceftizoxime, ceftazidime). Other examples include, but are not limited to Bensal HP, Barri-Care, Care-Creme, Formula Magic, Baza, Micro-Guard, Ca-Rezz, Diabet-X products, Mitrazol Powder, PiercingCare, Triple Care products, and various antifungal creams and powders.
g) Compression
[0225] Dynamic compression examples, include pumps and sleeves such as but not limited to ArtAssist, ArterialFlow, EdemaFlow, PulStar, Circulator Boot, Flowplus, Flowpress, Flowtron. Static compression include but are not limited to leg wrappings, gloves, socks, leg wears, leg supports, arm sleeves, stasis pads, compression hosieries, non-elastic bands, high compression bandages, zinc impregnated bandages, elastic bandages.
h) Oxygen Therapy
[0226] Examples of systemic hyperbaric oxygen therapy include but are not limited to compartments for one patient to lay down, for one patient to sit up to 25 degree angle, for one patient to sit up to 90 degree angle, for more than one patient to be treated simultaneously. Examples of topical hyperbaric oxygen therapy include but are not limited to disposable topical hyperbaric oxygen systems for extremity ulcers, disposable topical hyperbaric oxygen systems for decubitis, post-op and trauma wounds.
i) Hydrotherapy, Electric Therapy
[0227] Examples include but are not limited to dry hydrotherapy machines; non- contact thermal wound care systems for use on partial- and full-thickness wounds that maintain warmth and humidity in the wound area; systems that provide non-thermal, pulsed high frequency, high peak power, electromagnetic energy to treat edema and pain in acute and chronic wounds; systems that use controlled, localized negative pressure and support for moist wound healing; pulsatile irrigators with controllable pressures below 15 psi for site-specific treatment of various wounds with variety of tips; various wound irrigation and whirlpool systems.
j) Nutritional Therapy Products
[0228] Examples include but are not limited to isotonic, high-protein, fiber- containing tube feedings to support wound healing; high-protein, cholesterol-free nutritional supplements.
k) Cohesives, Glues, Sealants, Patches
[0229] Examples include but are not limited to Dermabond, CoStasis, CoSeal, BioGlue, FibRx, FocalSeal, FloSeal, AutoSeal, Indermil, Syvek, LiquiSheild, LiquiBand, Quixil, CryoSeal, VIGuard Fibrin Sealant, and various tapes, closures, and securement products.
1) Topical Wound Healing Promoters
[0230] Examples include but are not limited to topical aerosols which stimulate the capillary bed of chronic wounds; skin protectants with zinc-nutrient formulations; topical gels to help scars feel softer and smoother; hydrophilic ointments that cleanse degraded proteins, promote healthy granulation, control local inflammation and reduce wound odors; oil-and- water wound dressing emulsions that selectively recruit macrophages.
m) Other Biotherapy agents
[0231] Adjuvant therapy may also be used in conjunction with the present invention. The use of adjuvants or immunomodulatory agents include, but are not limited to tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-I, MIP-I beta, MCP-I, RANTES, and other chemokines.
2. Psoriasis
[0232] In accordance with the present invention, a lactoferrin composition provided in any of the above-described pharmaceutical carriers is orally, topically, or parenterally administered to a subject suspected of or having psoriasis.
[0233] Psoriasis is a chronic inflammatory skin disorder that undergoes repeated relapse and remission and affects one to three percent of the world's population. The disease can begin at any age and has its peak appearance in the third decade. Its severity, course, and remissions are unpredictable. (See, T. P. Habif, in: Clinical Dermatology, Mosby, Mo. (1996) and The Textbook of Medicine edts: J. B. Wyngaarden and L. H. Smith, W., W. B. Saundres Company, Philadelphia (1985)).
[0234] Patients with psoriasis can be divided into two groups, those with a genetic predisposition and those that respond to epigenetic, i.e. environmental factors. The most common form is chronic plaque psoriasis and is characterized by hyperplasia of the epidermis and inflammation of the dermis and epidermis. These changes arise due to activation of a T- lymphocyte cell mediated dermal immune reaction in regional lymph nodes in response to unidentified antigenic stimuli. The activated T-cells cause keratinocyte cells to proliferate and assume a psoriasis phenotype. The inflammatory reaction is caused by proinflammatory cytokine proteins that are induced in response to the environmental stimulus and results in infiltration of the dermis and epidermis by inflammatory white blood cells.
[0235] The present invention provides a method of enhancing the immune system of a subject suffering from psoriasis by administering to the subject a lactoferrin composition. Depending upon the mode of administration, different arms of the immune system are enhanced. or example, topical administration of the composition resulfs in" enhancement of the local immune system, i.e., in the vicinity of the wound. Parenteral administration of the composition results in enhancement of the systemic immune system. Yet further, oral administration of the composition results in enhancement of the mucosal immune system, which can also result in systemic effects as well.
[0236] Psoriasis has been shown to be caused by inappropriate local immune responses which involve the de novo production of TNF-α by keratinocytes (Gilhar et al., 1996, Clin. Exp. Immunol. 106: 134-142). Accordingly, in one embodiment, the present invention provides compositions that directly interfere with the production of TNF- α by keratinocytes, thus preventing the inappropriate local immune response causing psoriasis. More specifically, the present invention provides novel pharmaceutical compositions comprising a lactoferrin composition for the treatment of psoriasis, hi a preferred embodiment, the compositions are formulated for topical application in about 0.5% to about 5% carrier. In alternative embodiments, the compositions are formulated for intradermal injections. [0237] In order to increase the effectiveness of the composition of the present invention, it may be desirable to combine the composition of the present invention with other agents effective in the treatment of psoriasis, such as anti-inflammatory agents. This process may involve administering the composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the lactoferrin composition and the other includes the second agent(s).
[0238] Alternatively, the composition of the present invention may precede or follow the other anti-inflammatory agent treatment by intervals ranging from minutes to weeks. In embodiments where the other anti-inflammatory agent and inventive composition are administered or applied separately to the anti-inflammatory, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and the inventive composition would still be able to exert an advantageously combined effect on the psoriasis.
[0239] Typical anti-inflammatory agents are topical agents, for example, but not limited to are corticosteroids (hydrocortisone and analogs). Systemic corticosteroids, for example, can induce prompt resolution of psoriatic lesions, but suppression requires ever- increasing doses.
C. Respiratory Disorders
[0240] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is orally administered to a subject suspected of or having a respiratory disorder. The respiratory disorder can be an allergic or non-allergic respiratory disorder.
[0241] According to the invention, an allergic or non-allergic respiratory disorder includes asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis and allergic rhinitis. In specific embodiments, the respiratory disorder is characterized by increased responsiveness of the trachea and bronchi to various stimuli, i.e., allergens, resulting in widespread narrowing of the airways.
[0242] The composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the obstruction of the airway or the hyperresponsiveness of the respiratory system to the allergen or other stimuli. The amount of lactoferrin composition may vary from about 1 mg to about 100 g. Preferably, the composition that is orally administered contains the range of 1 mg to 10 g of lactoferrin composition per day, more preferably, 10 mg to about 1 g. More preferably, the composition of the present invention also contains metal chelators. A preferred metal chelator is EDTA. More preferably, the composition that is orally administered contains the ratio of 1:10,000 to about 2:1 EDTA to lactoferrin.
[0243] A further embodiment is a method of enhancing a mucosal immune response in the gastrointestinal tract in a subject comprising the step of administering orally to said subject the composition of the present invention. It is envisioned that oral administration of lactoferrin composition stimulates IL- 18 in the lungs, sinuses or systemically. Yet further, it is contemplated that an enhancement of lactoferrin composition can reduce the infiltration of inflammatory cells into the lung.
[0244] In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine these compositions with an additional agent. For example, known asthma agents are used in combination with the composition of the present invention. Exemplary agents known to treat asthma include mast cell degranulation agents (i.e., Cromylyn sodium or Nedocromil sodium), leukotriene inhibitors (i.e., Monteleukast sodium, Zafirlukast, or Pranlukast hydrate), corticosteroids (i.e., Beclomethasone, Budesonide, Ciclesonide, Hydrolysable glucocorticoid, Triamcinolone acetonide, Flunisolide, Mometasone furoate, or Fluticasone propionate), β-Antagonists (i.e., Albuterol, Bambuterol, Formoterol, Salbutamol, Terbutaline sulfate, or Salmeterol), IgE binding inhibitors (i.e., Omalizumab), Adenosine A2 agonists, Anti-CD23 antibody, E-Selectin antagonists, P-Selectin antagonists, L- Selectin antagonists, interlukin inhibitors/monoclonal antibodies, pulmonary surfactants, neurokinin antagonists, NF-Kappa-B inhibitors, PDE-4 inhibitors (i.e., Cilomilast, or Roflumilast), Thromboxan A2 inhibitors (i.e., Ramatroban, or Seratrodast), tryptase inhibitors, VIP agonists or antisense agents. [0245] Yet further, the agent is a known agent used to treat allergic rhinitis. Exemplary agents include, but are not limited to Hl antihistamines i.e., terfendine or astemizole; alpha-adrenergic agents; and glucocorticoids, i.e., beclamethasone or flunisolide.
[0246] In addition, the agent is a known agent to treat sinusitis, more specifically, chronic sinusitis. Exemplary agents/therpies include, but are not limited to surgery (e.g., enlarging a sinus passage, remove obstructing bone or nasal polyps, mucosal stripping, removal of sinuses); corticosteroids (e.g., oral, intranasal, nebulized, or inhaled); antibiotics (e.g., oral, intranasal, nebulized, inhaled or intravenous); anti-fungal agents; salt-water nasal washes and mist sprays; anti-inflammatory agents; decongestants (oral or nasal); guaifenesin; potassium iodide; leukotriene inhibitors (e.g., monteleukast); mast cell degranulating agents; topical moisterizing applications (e.g., nasal sprays or gels which may contain moisterizing agents such as propylene glycol or glycerin); hot air inhalation; mechanical devices to aid in breathing; enzymatic cleansers (e.g., papaya enzymes); and antihistame sprays.
[0247] Still further, the agent is a known agent to treat COPD or chronic bronchitis or emphysema. Exemplary agents/interventions include, but are not limited to oxygen; brbnchodilator drugs [e.g., short and long acting beta-2 stimulants, anticholinergics (e.g., ipratoprium bromide, theophylline compounds or a combination), steroids (topical or oral), or mucolytic agents (e.g., ambroxol, ergosterin, carbocysteine, iodinated glycerol)]; antibiotics; anti-fungals; moisterization by nebulization; anti-tussives; respiratory stimulants (e.g., doxapram, almitrine bismesylate); surgery (e.g., bullectomy, lung volume reduction surgery, lung transplantation); and alpha 1 antitrypsin administration.
[0248] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism. [0249] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination.
D. Infectious Diseases
[0250] In certain embodiment of the invention, the present invention is useful for the treatment and/or prevention of any inflamatory condition that is caused by an infectious disease. Infectious diseases include infections of viral etiology such as HIV, West Nile Virus, influenza, Herpes, viral hepatitis, Epstein Bar, polio, viral encephalitis, measles, chicken pox, Papilloma virus etc.; or infections of bacterial etiology such as bacterial meningitis, paramenigeal infections, septic thrombophlebitis, pneumonia, tuberculosis, myocarditis, bacteremia, syphilis, etc.; or infections of parasitic etiology such as malaria, trypanosomiasis, leishmaniasis, candidiasis, trichomoniasis, amoebiasis, etc.
[0251] Examples of infectious inflammatory conditions include but are not limited to pathogens such as meningococci, Salmonella typhi, Dengue virus, Plasmodia, Pneumocystis carinii, Yersinia enterocolitica pathogens, enterovirus, fungi.
1. Viral
[0252] The present invention can have applications therefore in the prevention and treatment of viral diseases. The following pathogenic viruses which are mentioned by way of example, influenza A, B and C, parainfluenza, paramyxoviruses, Newcastle disease virus, respiratory syncytial virus, measles, mumps, adenoviruses, adenoassociated viruses, parvoviruses, Epstein-Barr virus (EBV), rhinoviruses, coxsackieviruses, echoviruses, reoviruses, rhabdoviruses, lymphocytic choriomeningitis, coronavirus, polioviruses, herpes simplex (HSV), human immunodeficiency viruses (HIV), cytomegaloviruses, papillomaviruses, human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), varicella-zoster, poxviruses, rubella, rabies, picornaviruses, rotavirus and Kaposi associated herpes virus.
2. Bacterial
[0253] In addition to the viral diseases mentioned above, the present invention is also useful in the prevention, inhibition, or treatment of bacterial infections. The following bacteria are mention by way of example, including, but not limited to, serotypes of pneumococci, streptococci such as S. pyogenes, S. agalactiae, S. equi, S. canis, S. bovis, S. equinus, S. anginosus, S. sanguis, S. salivarius, S. mitis, S. mutans, other viridans streptococci, peptostreptococci, other related species of streptococci, enterococci such as Enterococcus faecalis, Enterococcus faecium, Staphylococci, such as Staphylococcus epidermidis, Staphylococcus aureus, particularly in the nasopharynx, Hemophilus influenzae, pseudomonas species such as Pseudomonas aeruginosa, Pseudomonas pseudomallei, Pseudomonas mallei, brucellas such as Brucella melitensis, Brucella suis, Brucella abortus, Bordetella pertussis, Neisseria meningitidis, Neisseria gonorrhoeae, Moraxella catarrhalis, Corynebacterium diphtheriae, Corynebacterium ulcerans, Corynebacterium pseudotuberculosis, Corynebacterium pseudodiphtheriticum, Corynebacterium urealyticum, Corynebacterium hemolyticum, Corynebacterium equi, etc. Listeria monocytogenes, Nocordia asteroides, Bacteroides species, Actinomycetes species, Treponema pallidum, Leptospirosa species and related organisms. The invention may also be useful against gram negative bacteria such as Klebsiella pneumoniae, Escherichia coli, Proteus, Serratia species, Acinetobacter, Yersinia pestis, Francisella tularensis, Enterobacter species, Bacteriodes and Legionella species and the like.
3. Fungal Infections
[0254] Yet further, either fungal and other mycotic pathogens or cells infected with fungal or other mycotic pathogens may be used in the present invention to prevent and/or treat diseases, rariging from mycoses involving skin, hair, or mucous membranes, such as, but not limited to, Aspergillosis, Black piedra, Candidiasis, Chromomycosis, Cryptococcosis, Onychomycosis, or Otitis externa (otomycosis), Phaeohyphomycosis, Phycomycosis, Pityriasis versicolor, ringworm, Tinea barbae, Tinea capitis, Tinea corporis, Tinea cruris, Tinea favosa, Tinea imbricata, Tinea manuum, Tinea nigra (palmaris), Tinea pedis, Tinea unguium, Torulopsosis, Trichomycosis axillaris, White piedra. Fungal and mycotic pathogens that can be used in the present invention include, but are not limited to, Absidia spp., Actinomadura madurae, Actinomyces spp., Allescheria boydii, Alternaria spp., Anthopsis deltoidea, Apophysomyces elegans, Arnium leoporinum, Aspergillus spp., Aureobasidium pullulans, Basidiobolus ranarum, Bipolaris spp., Blastomyces dermatitidis, Candida spp., Cephalosporium spp., Chaetoconidium spp., Chaetomium spp., Cladosporium spp., Coccidioides immitis, Conidiobolus spp., Corynebacterium tenuis, Cryptococcus spp., Cunninghamella bertholletiae, Curvularia spp., Dactylaria spp., Epidermophyton spp., Epidermophyton βoccosum, Exserophilum spp., Exophiala spp., Fonsecaea spp., Fusarium spp., Geotrichum spp., Helminthosporium spp., Histoplasma spp., Lecythophora spp., Madurella spp., Malassezia furfur, Microsporum spp., Mucor spp., Mycocentrospora acerina, Nocardia spp., Paracoccidioides brasiliensis, Penicillium spp., Phaeosclera dematioides, Phaeoannellomyces spp., Phialemonium obovatum, Phialophora spp., Phoma spp., Piedraia hortai, Pneumocystis carinii, Pythium insidiosum, Rhinocladiella aquaspersa, Rhizomucor pusillus, Rhizopus spp., Saksenaea vasiformis, Sarcinomyces phaeomuriformis, Sporothrix schenckii, Syncephalastrum racemosum, Taeniolella boppii, Torulopsosis spp., Trichophyton spp., Trichosporon spp., Ulocladium chartarum, Wangiella dermatitidis, Xylohypha spp., and Zygomyetes spp.
[0255] In addition, the invention may prove useful in controlling protozoan or macroscopic infections by organisms such as Cryptosporidium, Isospora belli, Toxoplasma gondii, Trichomonas vaginalis, Cyclospora species, for example, and for Chlamydia trachomatis and other Chlamydia infections such as Chlamydia psittaci, or Chlamydia pneumoniae, for example.
4. Bacteremia and septic shock
[0256] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having bacteremia, sepsis, septic shock or sequelae. These conditions could be caused by gram- negative, gram-positive bacteria or other infectious agents such as Candida in any foci of the body and are at a risk of developing into or have developed into a systemic inflammatory response syndrome.
[0257] Yet further, bacteremia may be caused by surgical manipulation of infected oral tissues or routine dental manipulations; catheterization of an infected lower urinary tract; incision and drainage of an abscess; and colonization of indwelling devices, especially IV and intracardiac catheters, urethral catheters, and ostomy devices and tubes. The primary site of infection is usually in the lungs, in the GU or GI tract, or in soft tissues including the skin in patients with decubitus ulcer. In chronically ill and immunocompromised subjects, gram- negative bacteremia occurs more commonly, than in a healthy subject. Additionally, these immunocompromised subjects may develop bloodstream infections caused by aerobic bacilli, anaerobes, and fungi. [0258] Predisposing factors for septic shock include diabetes mellirus; cirrhosis; leukopenic states, especially those associated with underlying neoplasms or treatment with cytotoxic agents; antecedent infection in the urinary, biliary, or GI tracts; invasive devices, including catheters, drainage tubes, and other foreign materials; and prior treatment with antibiotics, corticosteroids, or ventilatory devices. Septic shock occurs more often in newborns, subjects > 35 yr, pregnant women, and those seriously immunocompromised by underlying diseases or iatrogenic complications of treatment.
[0259] According to the invention, the above-described method is used for the prophylaxis of bacteremia, sepsis, septic shock, related conditions or their consequences. In specific embodiments, the disorder is characterized by a risk of endotoxemia resulting from the use of antibiotic and the subsequent release of endotoxin, as well as positively identified bacteremia.
[0260] A person at risk for developing bacteremia, sepsis, septic shock and/or related conditions is a person that is considered to be immunocompromised and/or chronically ill. The immunocompromised subject, who is, at the time of bacterial exposure, has a preexisting condition that reduces one or more mechanisms for normal defense against infection. The immunocompromised condition may be due to a defect or dysfunction of the immune system or to other factors that heighten susceptibility to infection, for example immunosuppressive agents. Thus, prophylatically, it is envisioned that the lactoferrin composition can reduce any of the following: the levels of circulating bacteria, the risk of the subject developing sepsis, septic shock, organ failure, and decrease the morbidity and mortality associated with bacteremia.
[0261] In a preferred embodiment of the present invention, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the risk of developing bacteremia and minimizing the effects of already existing bacteremia, sepsis, septic shock or related conditions. The amount of the lactoferrin in the composition may vary from about 1 mg to about 100 g.
[0262] The guiding principle in the use of the lactoferrin composition is to administer the treatment at the earliest signs of bacteremia, sepsis or septic shock being developed to attenuate the development of bacteremia and to reduce the extent of organ damage that results from sepsis and septic shock. The improvement is any observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease. In certain aspects, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate levels of bacteria in circulation. In further aspects, an improvement can consist of any of the following, for example, decrease in the levels of circulating bacteria, attenuating the development of sepsis, attenuating the development of septic shock, attenuating the development of organ failure, decreasing morbidity associated with bacteremia and decreasing mortality (death) associated with bacteremia. Thus, after administration of the lactoferrin composition, if any of the above conditions improve, then the amount of the lactoferrin composition is considered to be an effective amount. Yet further, administration of the lactoferrin composition will also attenuate the development of sepsis, septic shock and other conditions related thereto.
[0263] In certain aspects, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the severity of sepsis or septic shock. In further aspects, an improvement can consist of any of the following, for example, decreasing mortality, decreasing morbidity, attenuating the development organ failure, decreasing days of hospitalization, decreasing or eliminating days of intensive care such as in an intensive care unit, decreasing or eliminating the use of supportive care such as a mechanical ventilator or decreasing the incidence of sequelae such as ARDS. Survival in patients with organ failure at baseline and prevention and reversal of organ failure are also evaluated. Thus, after administration of the lactoferrin composition, if any of the above conditions improve, then the amount the lactoferrin composition are considered to be an effective amount.
[0264] In certain aspects, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate the severity of ALI or ARDS. In further aspects, an improvement can consist of any of the following, for example, decrease in mortality, attenuating the development organ failure, decreasing days of hospitalization, decreasing or eliminating days of intensive care such as in an intensive care unit, or decreasing or eliminating the use of supportive care such as a mechanical ventilator or PaO2ZFiO2 ratios. Thus, after administration of the lactoferrin composition, if any of the above conditions improve, then the amount of are considered to be an effective amount. [0265] In order to increase the effectiveness of the composition, it may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment or prevention of bacteremia, sepsis, septic shock and related conditions, for example known agents to treat bacterial infections, e.g., antibiotics, known agents for the treatment of sepsis, e.g., Drotrecogin alfa (activated) and agents to treat inflammation. In some embodiments, it is contemplated that a conventional therapy or agent, including but not limited to, a pharmacological therapeutic agent may be combined with the composition of the present invention.
[0266] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0267] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware -that the composition of the present invention and agents can be administered in any order or combination. In other aspects, one or more agents may be administered substantially simultaneously, or within about minutes to hours to days to weeks and any range derivable therein, prior to and/or after administering the composition.
[0268] Administration of the composition to a cell, tissue or organism may follow general protocols for the administration of cardiovascular therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
[0269] Pharmacological therapeutic agents and methods of administration, dosages, etc. are well known to those of skill in the art (see for example, the "Physicians Desk Reference", Goodman & Gilman's "The Pharmacological Basis of Therapeutics", "Remington's Pharmaceutical Sciences", and "The Merck Index, Eleventh Edition", incorporated herein by reference in relevant parts), and may be combined with the invention in light of the disclosures herein. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject, and such individual determinations are within the skill of those of ordinary skill in the art.
[0270] Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include an antimicrobial agent, an anti-sepsis agent, an antiinflammatory agent, an antithrombotic/fibrinolytic agent, a blood coagulant, an antiarrhythmic agent, an antihypertensive agent, a vasopressor, or agents to treat metabolic acidosis. In certain aspects of the present invention, antimicrobial agents, e.g., antibiotics are used in combination with the composition of the present invention.
a) Anti-microbial agents
[0271] In certain embodiments, an "antimicrobial agent" can be used in combination with the lactoferrin composition. An antimicrobial agent may comprise an antibiotic, anti-fungal, and anti-viral agent.
[0272] Antibiotics inhibits the growth of microorganisms without damage to the host. For example, the antibiotic may inhibit cell wall synthesis, protein synthesis, nucleic acid synthesis, or alter cell membrane function. Classes of antibiotics that can possibly be used in conjunction with the peptide include, but are not limited to, macrolides (i.e., erythromycin), penicillins (i.e., nafcillin), cephalosporins (i.e., cefazolin), carbepenems (i.e., imipenem, aztreonam), other beta-lactam antibiotics, beta-lactam inhibitors (i.e., sulbactam), oxalines (i.e. linezolid), aminoglycosides (i.e., gentamicin), chloramphenicol, sufonamides (i.e., sulfamethoxazole), glycopeptides (i.e., vancomycin), quinolones (i.e., ciprofloxacin), tetracyclines (i.e., minocycline), fusidic acid, trimethoprim, metronidazole, clindamycin, mupirocin, rifamycins (i.e., rifampin), streptogramins (i.e., quinupristin and dalfopristin) lipoprotein (i.e., daptomycin), polyenes (i.e., amphotericin B), azoles (i.e., fluconazole), and echinocandins (i.e., caspofungin acetate). Examples of specific antibiotics that can be used include, but are not limited to, erythromycin, nafcillin, cefazolin, imipenem, aztreonam, gentamicin, sulfamethoxazole, vancomycin, ciprofloxacin, trimethoprim, rifampin, metronidazole, clindamycin, teicoplanin, mupirocin, azithromycin, clarithromycin, ofloxacin, lomefloxacin, norfloxacin, nalidixic acid, sparfloxacin, pefloxacin, amifloxacin, gatifloxacin, moxifloxacin, gemifloxacin, enoxacin, fleroxacin, minocycline, linezolid, temafloxacin, tosufloxacin, clinafloxacin, sulbactam, clavulanic acid, amphotericin B, fluconazole, itraconazole, ketoconazole, and nystatin. Other examples of antibiotics, such as those listed in Sakamoto et al, U.S. Pat. No. 4,642,104 herein incorporated by reference will readily suggest themselves to those of ordinary skill in the art.
[0273] Anti-viral agents can also be used in combination the lactoferrin composition to treat and/or prevent a viral infection or disease. Such anti-viral agents include, but are not limited to protease inhibitors (e.g., saquinavir, ritonavir, amprenavir), reverse transcriptase inhibitors (e.g., azidothymidine (AZT), lamioridine (3TC), dideoxyinosine (ddl)), dideoxycytidine (ddC), zidovudine), nucleoside analogs (e.g., acyclovir, penciclovir).
[0274] In certain embodiments, anti-fungal agents can be used in combination with the peptide compositon to treat and/or prevent a fungal infection. Such anti-fungal agents include, amphotericin B (Amphocin®, Fungizone®), butoconazole (Femstat®), clotrimazole (Mycelex®, Gyne-Lotrimin®, Lotrimin®, Lotrisone®), fluconazole (Diflucan®), flucytosine (Ancobon®), griseofulvin (Fulvicin P/G®, Grifulvin V®, Gris-PEG®), itraconazole (Sporanox®), ketoconazole (Nizoral®), miconazole (Femizol-M®, Monistat®), nystatin (Mycostatin®), terbinafine (Lamisil®), terconazole (Terazol®), or tioconazole (Vagistat®).
b) Other agents
[0275] Anti-sepsis agents include, but are not limited to Drotrecogin alfa (activated). Agents used for the treatment of ALI and ARDS include but are not limited to intra- pulmonary instillation of surfactants, and leukotriene modifiers. Anti-inflammatory agents include, but are not limited to non-sterodial anti-inflammatory agents (e.g., naproxen, ibuprofen, celeoxib) and sterodial anti-inflammatory agents (e.g., glucocorticoids).
[0276] Non-limiting examples of non-pharmacologic interventions that may be used in the present invention include supportive care such as organ support in sepsis and septic shock and low tidal volume ventilation protocols in ALI and ARDS. E. Pain
[0277] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced pain which may be acute or chronic in nature.
[0278] Causes of acute pain include, but are not limited to pain following trauma such as a blunt or sharp injury, bullet wounds, or surgery.
[0279] Causes of chronic pain include, but are not limited to pain associated with chronic diseases such as cancer, arthritis, inflammatory disease, chronic wounds, cardiovascular accidents, disorders of the spinal chord or central nervous system, and recovery from surgery.
[0280] In a preferred embodiment of the present invention, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate pain. Thus, a subject is administered a therapeutically effective amount of a lactoferrin composition so that the subject has an improvement in the parameters relating to pain including subjective measures such as pain scores and grading and objective measures including use of additional pain medications. The improvement is any observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the patient condition, but may not be a complete cure of the disease. The amount of lactoferrin in the composition may vary from about 1 ng to about 100 g each day.
[0281] In certain embodiments, the lactoferrin related lactoferrin composition is administered to a subject to modulate chronic or acute pain in the subject. Modulation of pain is achieved by administering an effective amount of the composition to decrease, reduce, inhibit or abrogate pain. Specifically, it is contemplated that lactoferrin stimulates or enhances cytokine production or activity. For example, lactoferrin can enhance the production or activity of TNF-α resulting in modulation of pain, acute or chronic, in the subject.
[0282] In order to increase the effectiveness of the composition, it may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment of pain both directly and indirectly. In some embodiments, it is contemplated that a conventional therapy or agent, including but not limited to, a pharmacological therapeutic agent, a non-pharmacological pain management techniques or a combination thereof, may be combined with the composition of the present invention.
[0283] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks, hi embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0284] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination. In other aspects, one or more agents may be administered within of from substantially simultaneously, about minutes to hours to days to weeks and any range derivable therein, prior to and/or after administering the composition.
[0285] Administration of the composition to a cell, tissue or organism may follow general protocols for the administration of pain therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
a) Pharmacological Therapeutic Agents
[0286] Pharmacological therapeutic agents and methods of administration, dosages, etc. are well known to those of skill in the art (see for example, the "Physicians Desk Reference", Goodman & Gilman's "The Pharmacological Basis of Therapeutics", "Remington's Pharmaceutical Sciences", and "The Merck Index, Eleventh Edition", incorporated herein by reference in relevant parts), and may be combined with the invention in light of the disclosures herein. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject, and such individual determinations are within the skill of those of ordinary skill in the art. [0287] Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include non-steroidal anti-inflammatory drugs (NSAIDS) (e.g., aspirin, indomethacin, ibuprofen, naproxen, acetaminophen, and ketoprofen); opioid analgesics (e.g., codeine, morphine, methadone, demerol, fentanyl, oxymorphone, and levarphanol), second generation NSAIDs (e.g., COX-2 inhibitors); and anti-depressant drugs (e.g., tricyclin antidepressants, sedatives, tranquilizers, hypnotics, anti-histamines, and amphetamines).
b) Non-Pharmacological Pain Techniques
[0288] In certain aspects, a therapeutic agent may comprise a non-pharmacological pain management techniques Non-pharmacological pain management techniques are well known to those of skill in the art, and may comprise, but are not limited to acupuncture or acupressure, local anesthesia/analgesia, regional anesthesia/analgesia (e.g., epidural or spinal analgesia/anesthesia), general anesthesia/analgesia (e.g., intravenous anesthetics or opioid pump) and chiropractic. Non-pharmacological pain-reduction agents include, but are not limited to devil's, capsaicin, menthold or L-phenylalanine.
F. Cardiovascular Diseases
[0289] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced or is at risk of developing cardiovascular disease. Risk factors include, but are not limited to elevated levels of cholesterol or CRP. One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce circulating levels of total cholesterol or triglycerides or cardiovascular inflammation.
[0290] Cardiovascular diseases and/or disorders include, but are not limited to, diseases and/or disorders of the pericardium, heart valves (e.g., incompetent valves, stenosed valves, Rheumatic heart disease, mitral valve prolapse, aortic regurgitation), myocardium (e.g., coronary artery disease, myocardial infarction, heart failure, ischemic heart disease, angina) blood vessels (e.g., hypertension, arteriosclerosis, aneurysm) or veins (e.g., varicose veins, hemorrhoids). In specific embodiments, the cardiovascular disease is atherosclerosis.
[0291] In specific embodiments of the present invention, the lactoferrin composition is administered to a subject suffering from or at risk for developing atherosclerosis. Thus, it is envisioned that the lactoferrin composition modulates or reduces the severity and/or incidence of atherosclerosis.
[0292] Prophylactic treatment can be administered to those subjects at risk for developing atherosclerosis. One risk factor is an atherogenic lipoprotein profile. For example, a ratio of serum cholesterol to high density lipoproteins of above 5:1 indicates a higher than average risk of developing atherosclerosis. Other factors indicating increased risk for atherosclerosis include a serum cholesterol level of above 240 mg/dl; a high density lipoprotein level below about 35 mg/dl; and a low density lipoprotein level above about 160 mg/dl.
[0293] Another embodiment includes treating a human subject with an elevated level of circulating total cholesterol or CRP according to the then medically established guidelines. It is contemplated that the composition of the present invention reduces or attenuates the levels of circulating total cholesterol, low density lipoproteins or very low density lipoproteins. It is contemplated that the composition of the present invention can interfere with how cholesterol enters the circulation either via absorption from food (exogenous pathway) or synthesis by the liver (endogenous pathway).
[0294] Another embodiment includes treating a human subject having dyslipedemia. Dyslipidemias are disorders of lipoprotein metabolism, including lipoprotein overproduction or deficiency. These disorders may be manifested by elevation of the serum total cholesterol, low-density lipoprotein (LDL) cholesterol and triglyceride concentrations, and a decrease in the high-density lipoprotein (HDL) cholesterol concentration. Thus, it is evisioned that the composition of the present invention can reduce or abboragate the effects of lipoprotein metabolism, such as dyslipidemia.
[0295] In a preferred embodiment of the present invention, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate cardiovascular disease. Thus, a subject is administered a therapeutically effective amount of a lactoferrin composition so that the subject has an improvement in the parameters relating to cardiovascular disease including circulating levels of total cholesterol, HDL, LDL, VLDL, trigylcerides and C- reactive protein (CRP). The amount of lactoferrin in the composition may vary from about 1 ng to about 20 g. Preferably, the composition that is orally administered contains the range of 0.5 g to 5 g of lactoferrin per day.
[0296] The improvement is any observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the patient condition, but may not be a complete cure of the disease. In certain aspects, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate excess amounts of cholesterol levels in circulation. A subject requires treatment for cholesterol levels based upon any of the following situations: LDL of 160 mg/ml or greater; LDL of 130-159 mg/ml and also have two or more cardiovascular risk factors; LDL of 100 mg/ml or greater in subjects with coronary heart disease (CHD); triglycerides of 200 mg/dl or higher; total cholesterol of 240 mg/dl or higher or HDL of less than 40 mg/dl. Thus, after administration of lactoferrin related compounds, if any of the above conditions improve, then the amount is considered an effective amount to decrease, reduce, inhibit or abrogate cholesterol levels in the circulation.
[0297] Another embodiment is a method of reducing vascular inflammation by administering the composition of the present invention. Vascular inflammation can be tied to a number of the underlying processes contributing to atherosclerosis which include endothelial dysfunction, vascular proliferation and matrix alteration. Recent studies have emphasized the involvement of inflammation in mediating all stages of atherosclerosis. Vascular inflammation is thought to be a consequence of damage to the vascular endothelium and may also involve the proliferation of vascular smooth muscle cells (vsmcs). One precursor of lesion development in humans may be focal accumulation of vsmcs within the intima. In early atherosclerosis, vsmcs may contribute to the development of the atheroma through the production of pro-inflammatory mediators such as monocyte chemoattractant protein 1 and vascular cell adhesion molecule, and through the synthesis of matrix molecules required for the retention of lipoproteins. Inflammation of the vascular endothelium and proliferation of vsmcs may also impact the stability of the plaque through the formation of a firm fibrous cap. Indeed, in lipid-laden lesions in which the fibrous cap is thin and weak, there is evidence of vsmc apoptosis, especially at the "shoulder" region, associated with inflammation. In addition, the local inflammatory milieu can induce expression of collagenase and inhibit expression of proteolytic inhibitors, thus rendering the fibrous cap weak and susceptible to rupture. Lactoferrin, having known anti-inflammatory properties, may thus serve to inhibit the underlying processes associated with the development of atherosclerosis.
[0298] In further embodiments, the composition may also reduce vascular spasms or vascular hyper-reactivity. Vascular spasms are a sudden, brief tightening of a blood vessel, which can temporarily reduce blood flow to tissues supplied by that vessel.
[0299] Still further, the composition may also promote endothelial integrity or healing. Endothelia are the layer of cells lining the blood vessels. Endothelial dysfunction most commonly refers to impairment of endothelium-dependent vasodilation and widespread abnormalities in endothelial integrity and homeostasis. It is believed that HDLs help maintain endothelial integrity, facilitate vascular relaxation, inhibit blood cell adhesion to vascular endothelium, reduce platelet aggregability and coagulation, and may favor fibrinolysis. The integrity or completeness of the endothelia lining of the vessels is important to preventing/treating the development of plaques and atherosclerosis. Thus, it is envisioned that the composition of the present invention will promote or modulate endothelial integrity or healing.
[0300] Yet further, another embodiment is a method of preventing a cardiovascular disease in a subject at risk for developing a cardiovascular disease comprising the step of administering to the subject a lactoferrin composition in an amount sufficient to result in prophylaxis of the cardiovascular disease in the subject. In preferred embodiments, the cardiovascular disease is atherosclerosis. It is envisioned that the composition not only possess therapeutic benefits for those subjects suffering from cardiovascular diseases, but also possess prophylactic properties for those subjects at risk for developing cardiovascular disease. A subject at risk may or may not be cognizant of their disease state or potential disease state and may or may not be aware that they are need of treatment. Thus, prophylatically, it is envisioned that the composition can reduce any of the following: the levels of circulating total cholesterol, low density lipoproteins (LDL), very low density lipoproteins (VLDL), levels of vascular inflammation, circulating C-reactive protein (CRP), triglycerides, and the proliferation of vascular smooth muscle cells in the subject. Yet further, the composition may also increase the levels of circulating high density lipoproteins (HDL). [0301] In order to increase the effectiveness of the composition, it may be desirable to combine these compositions and methods of the invention with a known agent effective in the treatment or prevention of cardiovascular disease or disorder, for example known agents to treat or prevent atherosclerosis. In some embodiments, it is contemplated that a conventional therapy or agent, including but not limited to, a pharmacological therapeutic agent, a surgical therapeutic agent (e.g., a surgical procedure) or a combination thereof, may be combined with the composition of the present invention.
[0302] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0303] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination. In other aspects, one or more agents may be administered substantially simultaneously, ΌΓ ivithin about minutes to hours to" "days to weeks and any range derivable therein, prior to and/or after administering the composition.
[0304] Administration of the composition to a cell, tissue or organism may follow general protocols for the administration of cardiovascular therapeutics, taking into account the toxicity, if any. It is expected that the treatment cycles would be repeated as necessary. In particular embodiments, it is contemplated that various additional agents may be applied in any combination with the present invention.
a) Pharmacological Therapeutic Agents
[0305] Pharmacological therapeutic agents and methods of administration, dosages, etc. are well known to those of skill in the art (see for example, the "Physicians Desk Reference", Goodman & Gilman's "The Pharmacological Basis of Therapeutics", "Remington's Pharmaceutical Sciences", and "The Merck Index, Eleventh Edition", incorporated herein by reference in relevant parts), and may be combined with the invention in light of the disclosures herein. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject, and such individual determinations are within the skill of those of ordinary skill in the art.
[0306] Non-limiting examples of a pharmacological therapeutic agent that may be used in the present invention include an antihyperlipoproteinemic agent, an antiarteriosclerotic agent, an anti-cholesterol agent, an anti-inflammatory agent, an antithrombotic/fibrinolytic agent, a blood coagulant, an antiarrhythmic agent, an antihypertensive agent, or a vasopressor. In certain aspects of the present invention, anti-cholesterolemic agents are used in combination with the composition of the present invention. Anti-cholesterol agents include but are not limited to HMG-CoA Reductase inhibitors, cholesterol absorption inhibitors, bile acid sequestrants, nicotinic acid and derivatives thereof, fibric acid and derivatives thereof. HMG-CoA Reductase inhibitors include statins, for example, but not limited to atorvastatin calcium (Lipitor®), cerivastatin sodium (Baycol®), fluvastatin sodium (Lescol®), lovastatin (Advicor®), pravastatin sodium (Pravachol®), and simvastatin (Zocor®). Agents known to reduce the absorption of ingested cholesterol include, for example, Zetia®. Bile acid sequestrants include, but are not limited to cholestyramine, cholestipol and colesevalam. Other anti-cholesterol agents include fibric acids and derivatives thereof (e.g., gemfibrozil, fenofibrate and clofibrate); nicotinic acids and derivatives thereof (e.g., nician, lovastatin) and agents that extend the release of nicotinic acid, for example niaspan. Anti-inflammatory agents include, but are not limited to non-sterodial anti-inflammatory agents (e.g., naproxen, ibuprofen, celeoxib) and sterodial anti-inflammatory agents (e.g., glucocorticoids).
b) Surgical Therapeutic Agents
[0307] In certain aspects, a therapeutic agent may comprise a surgery of some type, which includes, for example, preventative, diagnostic or staging, curative and palliative surgery. Surgery, and in particular a curative surgery, may be used in conjunction with other therapies, such as the present invention and one or more other agents.
[0308] Such surgical therapeutic agents for cardiovascular diseases and disorders are well known to those of skill in the art, and may comprise, but are not limited to, performing surgery on an organism, providing a cardiovascular mechanical prostheses, angioplasty, coronary artery reperfusion, catheter ablation, providing an implantable cardioverter defibrillator to the subject, mechanical circulatory support or a combination thereof. Non-limiting examples of a mechanical circulatory support that may be used in the present invention comprise an intra-aortic balloon counterpulsation, left ventricular assist device or combination thereof.
G. Autoimmune Disorders
[0309] According to the present invention, a subject that is suspected of an autoimmune disease or a subject suffering from an autoimmune disease is treated with the lactoferrin composition of the present invention.
[0310] The pathogenesis of autoimmune disorders is mediated by the aberrant activation, differentiation and trafficking of leukocytes in response to tissue self-antigens. Autoimmune reactions can often have fatal consequences. They can cause the destruction of vital tissue which manifests in the development of chronic autoimmune diseases. The driving forces in the progression of these diseases are autoreactive immune cells. In most cases, these autoreactive cells are CD4 + T-cells which somehow escaped the self-tolerance control mechanisms of the immune system. In non-pathogenic situations, CD4+ T-cells act as T helper cells, which control or mediate the activation and differentiation of other immune cells such as cytotoxic CD 8+ T- cells, NK cells, granulocytes, macrophages and B-cells. However, in the situation of chronic autoimmune diseases, CD4 + T- cells are no longer just mediators, but are rather key players of the autoimmune response. In particular, they are either directly or indirectly responsible for the characteristic tissue destruction that occurs in autoimmune diseases, which is triggered by the recognition of autoantigens. These autoantigens are derived from self-proteins of the attacked tissue and are presented to the autoreactive CD4+ T- cells by antigen-presenting cells (APCs), such as macrophages, dendritic cells or B-cells. In addition, aberrant T cell-dependent or T cell- independent antibody production by activated B cells plays a critical role in initiating and amplifying tissue damage in autoimmune disorders.
[0311] Such autoimmune disorders that can be treated using the composition of the present invention include, but are not limited to, the following: Alopecia Areata, Ankylosing Spondylitis, Antiphospholipid Syndrome, aplastic anemia, myelodysplastic syndromes, paroxysmal nocturnal hemoglobulinemia, pure red cell aplasia, chronic neutropenias, amegakaryocytic thrombocytopenia, antiphospholipid syndromes, autoimmune thrombocytopenia, autoimmune hemolytic syndromes, antiphospholipid syndromes, autoimmune gastritis, achlorhydria, Autoimmune Addison's Disease, Autoimmune Diabetes, Autoimmune Hemolytic Anemia, Autoimmune Hepatitis, Autoimmune hypophysitis, Autoimmune orchiditis, autoimmune ovarian failure, Behcet's Disease, Bullous Pemphigoid, Cardiomyopathy, Celiac Sprue-Dermatitis, Cicatrical pemphigoid, Chronic Fatigue Immune Dysfunction Syndrome (CFIDS), Chronic Inflammatory Demyelinating Polyneuropathy, Interstitial cystitis, Churg- Strauss Syndrome, Cicatricial Pemphigoid, CREST Syndrome, Cold Agglutinin Disease, Crohn's Disease, Dermatitis herpetiformis, Discoid Lupus, Drug-induced autoimmune disorders, Endometriosis, Epidermolysis bullosa acquisita, Essential Mixed Cryoglobulinemia, Fibromyalgia-Fibromyositis, Glomerulonephritis, Good Pasture Syndrome, Graft Versus Host Disease, Graves' Disease, Guillain-Barre, Hashimoto's Thyroiditis, Idiopathic Inflammatory Myopathies, Idiopathic Pulmonary Fibrosis, Idiopathic Thrombocytopenia Purpura (ITP), IgA Nephropathy, Insulin Dependent Diabetes, Juvenile Arthritis, Lichen Planus, Systemic Lupus Erythmatosus, Meniere's Disease, Metal-induced autoimmunity disorders, Mixed Connective Tissue Disease, Multiple Sclerosis, Myasthenia Gravis, Myocarditis, Myositis, Optic neuritis, Painless/postpartum thyroiditis, Peripheral nerve vasculitis, Pemphigus Foliaceus, Pemphigus Vulgaris, Pernicious Anemia, Polyarteritis Nodosa, Polychondritis, Polyglandular Syndromes, Polymyalgia Rheumatica, Polymyositis and Dermatomyositis, Postinfectious autoimmune disorders, Primary Agammaglobulinemia, Primary Biliary Cirrhosis, Psoriasis, Psoriatic Arthritis, Reactive Arthritis, Raynaud's Phenomenon, Reiter's Syndrome, Rheumatic Fever, Rheumatoid Arthritis, Sarcoidosis, Scleritis, Scleroderma, Sjogren's Syndrome, Stiff-Man Syndrome, Takayasu Arteritis, Temporal Arteritis/Giant-cell Arteritis, Ulcerative Colitis, Uveitis, Vasculitis, Vitiligo, and Wegener's Granulomatosis.
[0312] In certain embodiments, the autoimmune disease that is treated includes, but is not limited to The present invention is also designed to treat any autoimmune disease including but not limited to Active Chronic Hepatitis, Addison's Disease, Anti-phospholipid Syndrome, Atopic, Allergy, Autoimmune, Atrophic Gastritis, Achlorhydra Autoimmune, Celiac Disease, Crohn's Disease, Cushings Syndrome, Dermatomyositis, (Type I) Diabetes, Discoid Lupus Erythematosis, Goodpasture's Syndrome, Grave's Disease, Hashimoto's Thyroiditis, Idiopathic Adrenal Atrophy, Idiopathic Thrombocytopenia, Insulin-dependent Diabetes, Lambert-Eaton Syndrome, Lupoid Hepatitis, Lymphopenia (some cases), Mixed Connective Tissue Disease, Multiple Sclerosis, Pemphigoid, Pemphigus Vulgaris, Pernicious Anema, Phacogenic Uveitis, Polyarteritis Nodosa, Polyglandular Auto. Syndromes, Primary Biliary Cirrhosis, Primary Sclerosing Cholangitis, Psoriasis, Raynauds, Reiter's Syndrome, Relapsing Polychondritis, Rheumatoid Arthritis, Schmidt's Syndrome, Scleroderma - CREST, Sjogren's Syndrome, Sympathetic Ophthalmia, Systemic Lupus Erythematosis, Takayasu's Arteritis, Temporal Arteritis, Thyrotoxicosis, Type B Insulin Resistance, Ulcerative Colitis, and Wegener's Granulomatosis
[0313] In certain embodiments, the composition of the present invention is administered to a subject suffering from graft versus host disease (GVHD), organ transplant rejection, autoimmune hepatitis, primary biliary cirrhosis, autoimmune cholangitis, primary sclerosing cholangitis, irritable bowel syndrome (IBS), multiple sclerosis (MS), chronic granulomatous disease, ankylosing spondylitis, scleroderma, polymyositis, (dermato)myositis, systemic vasculitis, systemic lupus erythematosus (SLE), Chrohn's disease, insulin-dependent diabetes (type 1) or ulcerative colitis.
1. Organ Transplant Rejection and GVHD
[0314] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is administered as a therapeutic drug to a subject _who has undergone organ transplant (kidney, heart, lung, liver, pancreas, bone marrow, ~ peripheral stem cells) and/or as a prophylactic drug to organ and/or tissue transplant, for example bone marrow (BM) or peripheral stem cell donor cells, to prevent the development of GVHD in recipients and/or to prevent or attenuate transplant rejection in the recipients. One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce chronic allograft rejection and toxicity associated with standard therapy, or the development of GVHD.
[0315] A further embodiment is treating, preventing or reducing the risk of developing graft-versus-host-disease by administering a lactoferrin composition to the donor organ or donor tissue prior to transplantation into the recipient. It is envisioned that administering the composition to the donor tissue/organ will attenuate the immune cells in the donor/organ and prevent the development of the immune response that is mounted against the recipient's tissue, thus preventing or attenuating GVHD. In further embodiments, the recipient and the donor organ/tissue can be treated with the composition of the present invention. [0316] Additional embodiments of the present invention include a method of treating, preventing or attenuating the severity of tissue or organ transplant rejection in a recipient comprising the step of administering to the donor a lactoferrin composition in an amount sufficient to attenuate the tissue or organ transplant rejection in the recipient. It is envisioned that the composition reduces allogeneic immune responses in the recipient. The composition can also be administered to both the recipient and the donor.
[0317] Another embodiment is a method of treating, preventing or attenuating the severity of xenograft tissue or xenograft organ transplant rejection in a recipient comprising the step of administering to the xenograft donor a composition in an amount sufficient to attenuate the tissue or organ transplant rejection in the recipient. The composition can also be administered to both the recipient and the xenograft donor.
[0318] In particular embodiments, progenitor cells or stem cells may be isolated from at least one organ, cell, tissue or organism. Stem cells can be isolated from embryonic or nonembryonic donors. The tissues from which the stem cells can be isolated include, for example, but are not limited to the bone marrow, the spleen, the liver, peripheral blood, umbilical cord tissue, umbilical cord blood, adipose tissue or skin. Yet further, tissue grafts may be used in the present invention. The tissue may be part or separated from an organism. In certain embodiments, a tissue may comprise, but is not limited to, adipocytes, alveolar, ameloblasts, axon, basal cells, blood (e.g., lymphocytes), blood vessel, bone, bone marrow, peripheral stem cells, brain, breast, cartilage, cervix, colon, cornea, embryonic, endometrium, endothelial, epithelial, esophagus, facia, fibroblast, follicular, ganglion cells, glial cells, goblet cells, kidney, liver, lung, lymph node, muscle, neuron, ovaries, pancreas, peripheral blood, prostate, skin, skin, small intestine, spleen, stem cells, stomach, testes, or ascite tissue.
[0319] In specific embodiments, an organ is the graft. Organs are comprised of tissues having a special function. Exemplary organs that are used in grafts in the present invention include, but are not limited to heart, kidney, pancreas, lung, or liver.
[0320] In a preferred embodiment of the present invention, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate chronic allograft rejection and toxicity related to standard therapy. The amount of lactoferrin in the composition may vary from about 1 ng to about 100 g, more preferably, lmg to about 2Og. Preferably, the composition that is orally administered contains the range of 0.1 g to 5 g of lactoferrin per day.
[0321] The improvement is any observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease. In certain aspects, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate levels of an immune response against a graft in the recipient. In further aspects, an improvement can consist of any of the following, for example, increased function of the graft, for example, increased urine output for kidney grafts or decreased jaundice for liver grafts; reduction in inflammation; reduction in general discomfort of the recipient; an overall increased tolerance for the graft. Thus, after administration of the lactoferrin composition, if any of the above conditions improve, then the amount is considered to be an effective amount.
[0322] An improvement in GHVD is also any observable or measurable improvement. Thus, one of skill in the art realizes that a treatment may improve the patient or subject's condition, but may not be a complete cure of the disease. In certain aspects, the composition is administered in an effective amount-to decrease, reduce, inhibit or abrogate levels of immune response from the donor's cells, tissue and/or organ against the host's tissues. GVHD can be acute or chronic or mild or severe. Improvements in acute symptoms include any of the following, for example, decrease skin rash, decrease diarrhea, increase in liver function, a decrease in susceptibility to infection. Improvements in chronic symptoms include, but are not limited to decrease skin rash, decrease dermatitis, decrease hair loss, a decrease liver damage, decrease dry eyes and mouth, a decrease susceptibility to infections and decrease lung and/or gastrointestinal disorders. Thus, after administration of the lactoferrin related composition, if any of the above conditions improve, then the amount is considered to be an effective amount.
[0323] Still yet, a further embodiment is a method of regulating a mucosal immune response in the gastrointestinal tract or a systemic immune response in a subject comprising the step of administering a lactoferrin related lactoferrin composition to said subject the composition of the present invention. It is envisioned that composition stimulates MIP-3 α and interleukin-18 in the gastrointestinal tract, which regulates immune responses. For example, interleukin-18 modulates both Thl/Th2 responses. It is known by those of skill in the art that IL- 18 plays an important role in allogeneic stem cell transplantation. Pre-treatment of allogeneic donors with IL- 18 significantly improves survival and reduces clinical and pathological indices of acute GVHD in BMT recipients. Other cytokines may also be enhanced or repressed for example, but not limited to IL-I5 IL-2, IL-6, ILlO, IL- 12 and GM-CSF. It is also envisioned that lactoferrin stimulates Thl/Th2 type-responses through the induction and/or repression of ThI and Th2 cytokines.
[0324] In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine these compositions with standard therapy. For example, known immunosuppressant agents are used in combination with the composition of the present invention. Exemplary agents known to prevent organ rejection are T cell modifiers such as cyclosporine (Neoral®, Sandimmune®), prednisone (Novo Prednisone®, Apo Prednisone®), azathioprine (Imuran®), tacrolimus or FK506 (Prograf®), mycophenolate mofetil (CellCept®), OKT3 (Muromorab CO3®, Orthoclone®), ATGAM® & Thymoglobulin® or serine-threonine phosphatase calcineurin (CN) inhibitors. In specific embodiments, the standard or approved treatment of GVHD, which is high doses corticosteroids, primarily high-dose methylprednisolone, is used in combination with the lactoferrin composition of the present invention.
[0325] In addition to immunosuppressant other anti-rejection and/or anti-GVHD therapies can be used in combination with the lactoferrin composition of the present invention. For example, therapies for preconditioning and prophylaxis of GVHD include, but are not limited to total body irradiation, cytosine arabinoside, L-phenylalanine mustard, cyclophosphamide, etoposide, triethylene thiophosphoramide, antithymocyte globulin, bisulfan, tacrolimus, methylprednisolone, cyclosporin, or methotrexate. Experimental therapies for treatment of GVHD include, but are not limited to cytokine inhibitors/antagonists (e.g., anti- TNFα antibody); IL-I receptor antagonist; recombinant IL-I receptor; inhibitors of T-cell activation (e.g., tacrolimus); antimetabolites (e.g., mycophenolate mofetil); anti-CD3 antibody (muromonab, OKT3); anti-CD25 antibody; anti-IL2 receptor monoclonal antibody daclizumab; extracorporeal photopheresis using ex vivo 8-methoxypsoralen; anti-thymocyte globulins (Thymoglobulin® or ATGAM® ); ABX-CBL or CBL-I; or Visilizumab (Nuvion®). [0326] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0327] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination.
H. Endocrine/Metabolism Disorders 1. Diabetes mellitus
[0328] In accordance with the present invention, the composition provided in any of the above-described pharmaceutical carriers is administered to a subject who has experienced or is at high risk of having diabetes mellitus. Thus, it is envisioned that the composition of the present invention may be administered to a subject to regulate diabetes mellitus. The composition modulates at least one symptom of diabetes mellitus, for example, decrease blood glucose or modulate blood insulin levels.
[0329] Risk factors for type I diabetes include islet-cell antibodies and those of type 2 or gestational diabetes include inactivity, obesity, siblings with diabetes, and history of diabetes during pregnancy. One of skill in the art can determine the patients who would potentially benefit from a therapeutic agent that would reduce circulating levels of glucose.
[0330] hi a preferred embodiment of the present invention, the composition is administered in an effective amount to decrease, reduce, inhibit or abrogate high glucose, or to reduce total body weight, glycosylated hemoglobin (HbAIc), or blood pressure or to modulate blood insulin levels. In the case of a diabetic condition, successful reduction of hyperglycemia by the lactoferrin composition may be manifested by the fasting plasma glucose level falling below 126 mg/dL, the 2-hour plasma glucose level during an oral glucose tolerance test (OGTT ) falling below below 200 mg/dL, or if a random plasma glucose determination reading below 200 mg/dL in a symptomatic individual. In the case of a pre-diabetic condition, a successful reduction of hyperglycemia by the lactoferrin composition may be manifested by the fasting plasma glucose falling below 110 mg/dL and/or the 2-hour plasma glucose on the OGTT falling below between 140 mg/dL. The amount of lactoferrin in the composition may vary from about 0.1 μg to about 100 g, more preferably, from about 1 μg to about 20 g, or any range therebetween. In specific embodiments, the composition that is administered contains the range of 0.1 g to 5 g of lactoferrin per day.
[0331] Glycohemoglobin (or glycosylated hemoglobin) is measured to monitor or accurately record blood glucose levels, and it is not influenced by acute changes in blood glucose or by the interval since the last meal. Glycohemoglobin is formed when glucose reacts non- enzymatically with the hemoglobin A molecule and is composed of several fractions, the major one being Hb AIc . Total glycohemoglobin (Hb Al) and Hb AIc (expressed as the percentage of total hemoglobin) vary in proportion to the average level of glucose over the lifespan of the red blood cell (RBC), thereby providing an index of glycemic control.
[0332] Further aspects of the invention include reducing blood glucose in a patient suffering from diabetes mellitus by administering to a subject an effective amount of the composition such that the amount of the lactoferrin composition modulates blood glucose. The blood glucose is monitored by the level of glycosylated hemoglobin (HbAIc).
[0333] Another aspect is a method of modulating blood insulin in a patient suffering from diabetes mellitus by administering to a subject an effective amount of the composition such that the amount modulates blood insulin. Modulating blood insulin includes reducing or maintaining blood insulin levels or increasing blood insulin levels. Modulating blood insulin levels in the present invention includes increasing blood insulin levels in insulin- dependent diabetes (Type I) or decreasing or maintaining insulin levels in non-insulin-dependent diabetes (Type II).
[0334] In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine the compositions with an additional agent. For example, known diabetes agents are used in combination with the composition of the present invention. Exemplary agents known to treat high glucose or insulin sulfonylureas, biguanides, alpha-glucosidase, thiazolidinedione, meglitinide, and amino acid D-phenylalanine derivative. Other antidiabetic agents may also include, a weight loss regimen and/or a diet alteration.
[0335] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0336] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination.
2. Treatment of Fragile Bone Diseases
[0337] In accordance with the present invention, the petpide composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a fragile bone condition.
[0338] The present invention is designed for the treatment of any type of fragile bone condition or any disorder relating to bone loss including but not limited to osteoporosis, age-associated osteoporosis, postmenopausal osteoporosis, osteitis deformans (Paget' s disease), osteogenesis imperfecta (brittle bones), and osteopetrosis.
[0339] Bone loss disorders arise from an imbalance in the formation of new healthy bone and the resorption of old bone, skewed toward a net loss of bone tissue. This bone loss includes a decrease in both mineral content and protein matrix components of the bone, and leads to an increased fracture rate of, predominantly, femoral bones and bones in the forearm and vertebrae. These fractures, in turn, lead to an increase in general morbidity, a marked loss of stature and mobility, and, in many cases, an increase in mortality resulting from complications.
[0340] Bone loss occurs in a wide range of subjects, including postmenopausal women, patients who have undergone hysterectomy, patients who are undergoing or have undergone long-term administration of corticosteroids, patients suffering from Cushing's syndrome, and patients having gonadal dysgensis.
[0341] Unchecked, bone loss can lead to osteoporosis, a major debilitating disease whose prominent feature is the loss of bone mass (decreased density and enlargement of bone spaces) without a reduction in bone volume, producing porosity and fragility.
[0342] Osteoporosis is common in the elderly of both sexes but is more pronounced in postmenopausal women. Osteoporosis may occur as a primary disorder or as a secondary complication of several diseases. It is proposed that genetic factors determine the size of the bone mass achieved in young adulthood. With aging, the increased osteoclastic function and the slowing of osteoblastic activity induced by endocrine influences, particularly decreased estrogen levels, result in a net negative balance in the continued turnover of bone. Osteoporosis causes bone pain owing to microfractures; results in loss in height and stability of the vertebral column; and predisposes to fractures of femoral necks, wrists, and vertebrae. The condition remains asymptomatic until skeletal fragility is well advanced.
[0343] Paget' s disease is currently considered to be a slow paramyxoviral infection of osteoblasts and then osteoclasts. The condition is divided into an initial osteolytic stage, followed by a mixed osteolytic-osteoblastic stage, evolving ultimately into burn-out quiescent osteosclerotic stage. Because new bone formation in active disease is disordered and poorly mineralized, it is soft and porous, lacks structural stability, and is vulnerable to fracture or deformation under stress. Patients may demonstrate fractures, nerve compression, osteoarthritis, and skeletal deformities.
[0344] Osteogenesis imperfecta or brittle bones refers to a group of closely related genetic disorders caused by qualitative or quantitative abnormal synthesis of type I collagen, constituting about 90% of the matrix of bone. Syndromes range from one variant (type II) that is uniformly fatal in the perinatal period (from multiple bone fractures) to other variants marked by increased predisposition to fracture but compatible with survival. Morphologically the basic change in all is osteopenia or too little bone, with marked thinning of the cortices and rarefication of the trabeculae. [0345] Osteopetrosis refers to a group of rare hereditary diseases characterized by overgrowth and sclerosis of boone, with marked thickening of the cortex and narrowing or filling of the medullary cavity impairing hematopoisesis. Despite too much bone, it is brittle and fractures easily. The autosomal recessive form is evident from birth, with anemia, neutropenia, infections and eventual death. The autosomal dominant form is benign but predisposes to fractures. Common to all forms is a hereditary defect in osteoclast function resulting in reduced bone resorption and enhanced net bone overgrowth.
[0346] In a preferred embodiment of the present invention, lactoferrin composition is administered in an effective amount to stabilize or reduce bone fragility, to strengthen the bone, inducing the growth of healthy bone, modulate calcium levels, and modulate mineral accumulation in the skeleton. The amount may vary from about 1 μg to about 100 g of lactoferrin composition. Preferably, the amount that is administered is in the range of 10 mg to 25 g. Most preferably, the amount that is administered is in the range of 10 μg to 5g.
[0347] It is often desirable to deliver the composition of the present invention to the subject over prolonged periods of time, for example, for periods of one week to one year from a single administration. Various slow release, depot or implant dosage forms may be utilized. For example, a dosage form a may contain a pharmaceutically acceptable non-toxic salt of the compounds which has a low degree of solubility in body fluids, for example, (a) an acid addition salt with a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene mono- or di-sulfonic acids, polygalacturonic acid, and the like; (b) a salt with a polyvalent metal cation such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed from e.g., N,N'-dibenzyl-ethylenediamine or ethylenediamine; or (c) combinations of (a) and (b) e.g. a zinc tannate salt. Additionally, the compounds of the present invention or, preferably, a relatively insoluble salt such as those just described, may be formulated in a gel, for example, an aluminum monostearate gel with, e.g. sesame oil, suitable for injection. Particularly preferred salts are zinc salts, zinc tannate salts, pamoate salts, and the like. Another type of slow release depot formulation for injection would contain the compound or salt dispersed for encapsulation in a slow degrading, non-toxic, non-antigenic polymer such as a polylactic acid/polyglycolic acid polymer for example as described in U.S. Pat. No. 3,773,919. The compounds or, preferably, relatively insoluble salts such as those described above may also be formulated in cholesterol matrix silastic pellets, particularly for use in animals. Additional slow release, depot or implant formulations, e.g. liposomes, are well known in the literature. See, for example, "Sustained and Controlled Release Drug Delivery Systems", J. R. Robinson ed., Marcel Dekker, Inc., N. Y., 1978.
[0348] Still yet, a further embodiment is a method of promoting the stimulation or inhibition of cytokines, growth factors or mediators of bone growth or resorption. Such growth factors include, but not limited to insulin-like growth factor- 1 (IGF-I), transforming growth factor-beta (TGF -b), and growth hormone releasing factor (GRF). Mediators of bone growth or resorption include, but are not limited to parathyroid hormone (PTH), calcitonin or 1,25- dihydroxycholecalciferol (1 ,25(OH)2D3).
[0349] Still yet, a further embodiment is a method of activating or inhibiting cells involved in bone remodeling including but not limited to osteoblasts, osteoclasts, chondrocytes, endothelial cells, and fibroblasts.
[0350] In order to increase the effectiveness of the composition of the present invention, it may be desirable to combine the composition of the present invention with other -agents effective in the treatment of fragile bone diseases including but not limited to calcium and cholecalciferol (vitamin D) supplements, estrogen replacement therapy (ERT), raloxifene, calcitonin, bisphosphonates such as alendronate and cyclical etidronate, statins, recombinant human parathyroid hormone (rhPTH), insulin-like growth factor- 1 (IGF-I), and transforming growth factor-beta (TGF-I). This process may involve administering the lactoferrin composition of the present invention and the agent(s) or multiple factor(s) at the same time. This may be achieved by administering a single composition or pharmacological formulation that includes both agents, or by administering two distinct compositions or formulations, at the same time, or at times close enough so as to result in an overlap of this effect, wherein one composition includes the human lactoferrin composition and the other includes the second agent(s).
[0351] Alternatively, the composition of the present invention may precede or follow the other bone healing agent treatment by intervals ranging from minutes to weeks. In embodiments where the other bone healing agent and the inventive composition are administered or applied separately, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and the composition would still be able to exert an advantageously combined effect on the condition.
I. Immunosuppressed Subjects
[0352] In order to carry out methods of the present invention, the composition of the present invention is administered to an immunosuppressed individual or an individual whose immune system is expected to be suppressed.
[0353] As noted above, the subject method is suited for preventing infection and/or inflammation in immunosuppressed individuals or individuals whose immune systems are expected to be repressed. In one aspect, the immunosuppressed subject exhibits neutropenia, or suffers from AIDS or any form of cancer including lymphoma and myeloma. In yet another aspect, the subject undergoes or is expected to undergo a therapy selected from the group consisting of chemotherapy, radiotherapy, and autologous peripheral stem cell transplant.
[0354] An effective amount of the lactoferrin composition depends on the severity and/or course of immunosuppression, the patient's clinical history and response, and the discretion of the attending physician. The composition is suitably administered to the patient at_ one time or over a series of treatments. The initial candidate dosage may be administered to a patient. The proper dosage and treatment regimen can be established by monitoring the progress of therapy using conventional techniques known to the people skilled of the art.
[0355] The amount of active ingredients that may be combined with the carrier materials to produce a single dosage form will vary depending upon the subject treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors, including the activity of the specific composition employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy, and can be determined by those skilled in the art.
[0356] In order to achieve effective prophylaxis, preferably, the lactoferrin composition is administered prior to the inception neutropenia, more specifically Grade 3 or higher neutropenia and continued on the same schedule daily for the entire period during which the patient is at a high risk of neutropenic fever, infection or mucositis.
[0357] It is envisioned that the immune response is enhanced by lactoferrin stimulating cytokines and/or chemokines. Exemplary cytokines include interleukin-18 and GM- CSF in the gastrointestinal tract, which are known to enhance immune cells or stimulate production of immune cells.
[0358] Certain treatment regimens include administering the composition in which the concentration of lactoferrin that is provided is in the range of 1 g to about 100 g/ ml and the composition is a solution that is swished in the mouths of the subjects for a few minutes (e.g., about 4 minutes) and then swallowed. This process can be repeated mutiple times a day, for example twice or more each day, for a total of three doses per 24 hour period. The doses are taken immediately after meals (one each after breakfast, lunch and dinner), or at similarly spaced intervals throughout the day. Subjects are asked not to eat or drink anything for a least one hour after swallowing the dose. The treatment may be terminated in the event of significant drug related adverse effects observed during one of the interim safety analyses.
[0359] The multiple administration of the lactoferrin composition each day can maximize the exposure of the oral cavity and gut to the drug. Preferably, patients an effective amount of the composition prophylactically for 21-28 days starting from the commencement of chemotherapy. Daily dosing continues through the neutropenic phase until the earliest of the following: a) Absolute Neutrophil Count (ANC) is > 600 cells/μL for three consecutive days, and mucositis (if present) has improved to Grade 2 or better for three consecutive days; or b) patient is discharged from the hospital or outpatient transplant setting. A last-day-of-therapy evaluation is performed within 24 hours after the last dose. The patients are also required to return for an end-of-treatment evaluation, to be performed 3-7 days after the last dose.
[0360] The treatment regimen in the present invention can prevent or attenuate infections in immunosuppressed patients, for example, patients undergoing aggressive chemotherapy or radiotherapy. The treatment reduces incidence, duration, and severity, and prolongs the infection-free interval of neutropenic infection (neutropenic fever). For the purpose of this invention, fever is considered as an oral temperature of at least 38.0°C on at least two
91 occasions within 24 hours, or a single oral temperature of at least 38.3°C. Neutropenic fever is defined as fever that begins when the patient is neutropenic (ANC < 500). The end of neutropenic fever is defined as the point at which the patient begins a period of 48 hours without fever (as defined above), whether or not the patient is still neutropenic. Incidence of neutropenic fever is calculated as the number of patients experiencing one or more episodes of neutropenic fever. Onset of neutropenic fever is determined in two ways: (1) days between the transfusion and the first episode of neutropenic fever and (2) days between a neutrophil count < 1000/uL and the first episode of neutropenic fever. Duration of neutropenic fever is defined as the number of days between the onset and the end of neutropenic fever. Severity of neutropenic fever is determined by the average temperature of the patient during the period of neutropenic fever. Infection-free interval is the period between the end of an infective fever and the onset of the next infective fever.
[0361] The present treatment also reduces the incidence, severity, and duration of oral and GI mucositis by improving all clinical criteria used for mucositis evaluation (pain, salivation, appetite, and oral inflammation). The incidence, duration, and severity of oral mucositis are assessed for each patient. Oral mucositis is graded using the Oral Mucositis Assessment Scale (OMAS). __ _ . - -
[0362] Prophylactic and concomitant use of antibiotics may be used during the treatment according to patient's conditions and usual standard of care in the medical profession. Use of hematopoietic growth factors is not recommended routinely, but G-CSF may be used as part of the standard of clinical carte at the discretion of the doctors. All medications necessary for the patient's well being may also be administered.
[0363] The responses of the patients in the course of treatment are closely monitored by evaluating various response parameters. The treatment protocol may be adjusted accordingly depending on the improvement of the patient's physical conditions.
J. Gastrointestinal Disorders
[0364] In accordance with the present invention, the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a gastrointestinal disorder. [0365] Gastrointestinal disorders include, but are not limited to, heartburn, bloating, postoperative ileus, abdominal pain and discomfort, early satiety, epigastric pain, nausea, vomiting, burbulence, regurgitation, intestinal pseudoobstruction, anal incontinence, gastroesophageal reflux disease, irritable bowel syndrome, ulcerative colitis, Crohn's disease, menstrual cramps, pancreatitis, spastic and interstitial cystitis and ulcers and the visceral pain associated therewith.
[0366] In certain embodiments, the lactoferrin composition is administered to a subject suffering from Crohn's disease, colitis, necrotizing entercolitis, endometriosis, irritable bowel syndrome, pancreatitis, periodontal disease, and ulcerative colitis.
K. Neurological Conditions
[0367] In accordance with the present invention, the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a neurological disorder.
[0368] It is envisioned that the composition of the present invention when " administered to a subject suffering from a neurological disorder, stimulates the immune system thereby alleviating the neurological conditions. Thus, the composition of the present invention can be useful with regard to the prevention, treatment, or amelioration of neurological, psychiatric, psychological, conscious state, behavioral, mood, and thought activity (unless otherwise indicated these will be collectively referred to herein as "neurological activity" which includes "psychological activity" or "psychiatric activity"). When referring to a pathological or undesirable condition associated with the activity, reference may be made to a neurological disorder which includes "psychiatric disorder" or "psychological disorder" instead of neurological activity or psychiatric or psychological activity. Although the activity to be modulated usually manifests itself in the form of a disorder such as a attention or cognitive disorders (e.g., Autistic Spectrum Disorders); mood disorder (e.g., major depressive disorder, bipolar disorder, and dysthymic disorder) or an anxiety disorder (e.g., panic disorder, posttraumatic stress disorder, obsessive-compulsive disorder and phobic disorder); neurodegenerative diseases (e.g., multiple sclerosis, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, sleep disorders, and chronic idiopathic demyelinating disease (CID)), one skilled in the art appreciates that the invention may also find application in conjunction with enhancing or diminishing any neurological or psychiatric function, not just an abnormality or disorder.
[0369] In certain embodiments, the lactoferrin related lactoferrin composition is administered to a subject suffering from multiple sclerosis, Alzheimer's disease, Parkinson's disease, muscular dystrophy, sleep or depression.
[0370] Neurological are diagnosed based upon the accumulation of physical, chemical, and historical behavioral data on each patient. One of skill in the art is able to perform the appropriate examinations to accumulate such data. One type of examination can include neurological examinations, which can include mental status evaluations, which can further include a psychiatric assessment. Other types of examinations can include, but are not limited to, motor examination, cranial nerve examination, and neuropsychological tests (e.g., Minnesota Multiphasic Personality Inventory, Beck Depression Inventory, or Hamilton Rating Scale for Depression). Thus, any improvement is based upon an improvement in any of the examinations that are used to diagnose a neurological disorder.
[0371] In addition to the above examinations, imaging techniques can be used to determine normal and abnormal brain function that can result in disorders. Functional brain imaging allows for localization of specific normal and abnormal functioning of the nervous system. This includes electrical methods such as electroencephalography (EEG), magnetoencephalography (MEG), single photon emission computed tomography (SPECT), (SPECT), as well as metabolic and blood flow studies such as functional magnetic resonance imaging (fMRI), and positron emission tomography (PET) which can be utilized to localize brain function and dysfunction.
[0372] In order to increase the effectiveness of administration of the composition of the present invention, it is desirable to combine the compositions with an additional agent. For example, known agents that are known to be stimulatory or inhibitory are used in combination with the composition of the present invention to treat a variety of neurological conditions. [0373] Herein, stimulating drugs comprise medications, anesthetic agents, synthetic or natural peptides or hormones, neurotransmitters, cytokines and other intracellular and intercellular chemical signals and messengers, and the like. In addition, certain neurotransmitters, hormones, and other drugs are excitatory for some tissues, yet are inhibitory to other tissues. Therefore, where, herein, a drug is referred to as an "excitatory" drug, this means that the drug is acting in an excitatory manner, although it may act in an inhibitory manner in other circumstances and/or locations. Similarly, where an "inhibitory" drug is mentioned, this drug is acting in an inhibitory manner, although in other circumstances and/or locations, it may be an "excitatory" drug.
[0374] Similarly, excitatory neurotransmitter agonists (e.g., norepinephrine, epinephrine, glutamate, acetylcholine, serotonin, dopamine), agonists thereof, and agents that act to increase levels of an excitatory neurotransmitter(s) (e.g., edrophonium; Mestinon; trazodone; SSRIs (e.g., flouxetine, paroxetine, sertraline, citalopram and fluvoxamine); tricyclic antidepressants (e.g., imipramine, amitriptyline, doxepin, desipramine, trimipramine and nortriptyline), monoamine oxidase inhibitors (e.g., phenelzine, tranylcypromine, isocarboxasid)), generally have an excitatory effect on neural tissue, while inhibitory neurotransmitters (e.g., dopamine, glycine, and gamm^-a^minobutyric_acid XGABA)), agonists thereof, ^and agents that act to increase levels of an inhibitory neurotransmitter(s) generally have an inhibitory effect. (Dopamine acts as an excitatory neurotransmitter in some locations and circumstances, and as an inhibitory neurotransmitter in other locations and circumstances.) However, antagonists of inhibitory neurotransmitters (e.g., bicuculline) and agents that act to decrease levels of an inhibitory neurotransmitter(s) have been demonstrated to excite neural tissue, leading to increased neural activity. Similarly, excitatory neurotransmitter antagonists (e.g., prazosin, and metoprolol) and agents that decrease levels of excitatory neurotransmitters may inhibit neural activity. Yet further, lithium salts and anesthetics (e.g., lidocane) may also be used in combination with the present invention.
[0375] In specific embodiments, antidepressants are administered in combination with the present invention to treat an affective disorder, such as depression. Such antidepressants include, but are not limited to monoamine oxidase inhibitors, 5-HT1A agonists, selective 5-HT uptake inhibitors, imipramine or clomipramine. [0376] The composition of the present invention may precede, be co-current with and/or follow the other agent(s) by intervals ranging from minutes to weeks. In embodiments where the composition of the present invention, and other agent(s) are applied separately to a cell, tissue or organism, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the composition and agent(s) would still be able to exert an advantageously combined effect on the cell, tissue or organism.
[0377] Various combination regimens of the composition and one or more agents are employed. One of skill in the art is aware that the composition of the present invention and agents can be administered in any order or combination.
L. Hematological Disorders
[0378] In accordance with the present invention, the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a hematological disorder. Such disorders can include, but are not limited to anaemia, sickel cell anemia or cachexia.
M. Ocular Disorders
[0379] In accordance with the present invention, the lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having an ocular disorder. Such disorders include, but are not limited to conjunctivitis, dry eye disease, glaucoma, allergic eye disease, uveitis or ocular infection.
[0380] Dry eye disease or keratoconjunctivitis sicca affects about 4 million people in the U.S. of which about 1.5 million have moderate to severe disease. Globally, 60 million people use artificial tears (Damato et al. 1984, Hikichi et al. 1995, Bjerrum et al. 1997, Schein et al. 1997). Despite the high levels of incidence, there is currently no highly effective treatment. Development of therapeutic treatments has been hampered by the lack of knowledge regarding the etiology of the condition to its complexity. However, it has now been suggested that inflammation affecting the lacrimal gland and ocular surface underlies the pathophysiological process (Pflugfelder et al. 1986, Raphael et al. 1988, Pepose et al. 1990, Kroemer et al. 1991, Jones et al. 1994, Pflugfelder et al. 1999, Turner et al. 2000). Thus, it is contemplated that the lacxoierrin composition ot the present invention can be used to decrease the inflammatory response thereby treating the ocular disease or disorder.
[0381] In certain embodiments, the lactoferrin compositions are preferably formulated as topical ophthalmic suspensions or solutions, with a pH of about 4 to 8. The establishment of a specific dosage regimen for each individual is left to the discretion of the clinicians. The amount of lactoferrin will normally be contained in these formulations in an amount 0.01% to 5% by weight, but preferably in an amount of 0.05% to 2% and most preferably in an amount 0. 1 to 1.0% by weight. The dosage form may be a solution, suspension microemulsion. Thus, for topical presentation 1 to 2 drops of these formulations would be delivered to the surface of the eye 1 to 4 times per day according to the discretion of a skilled clinician.
N. Fibrotic Diseases
[0382] The present invention is also designed to treat any fibrotic condition including but not limited to diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis (cirrhosis), and atherosclerosis.
O. Non-Infectious related Inflammatory conditions
[0383] The present invention is also designed to treat any inflammatory condition, being non-infectious. Examples of non-infectuious inflammatory conditions include but are not limited to rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ specific inflammatory diseases, glomerular disease, Sjogrens disease, or Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
IV. Examples
[0384] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1 Tests for chemotactic modulators
[0385] The migration of cells towards a source of a chemotactic modulator is tested in a chemotaxis or chemoattraction cell migration assay, including lactoferrin, chemokines, chemokine receptors, inflammatory or immune mediators, viral anti-chemokines, a mixture of casein and autologous serum, chemotactic C 5 fragment, and formyl-1-methionyl-l-leucine-l- phenylalanine (f-Met-Leu-Phe), bacterial factor derived from Escherichia coli, the simple peptide formylmethionylalanine, and activated human complement, carbamylcholine, phenylephrine, and cyclic guanosine 5 '-monophosphate, nucleosides and nucleotides, and prostaglandins. Chemotactic modulators are tested alone or in combination with other chemotactic modulators to assess additive, synergistic, agonistic or antagonistic effects.
[0386] The different cell types used are deriyeji from healthy or diseased tissue and are from human, mouse or other species, including T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells. Cells from wild type or knockout mice are used to determine the mechanisms behind the chemotactic modulation.
Example 2 Lactoferrin acts as a chemokine, a chemokine blocker or a chemokine enhancer
[0387] The migration of human monocytic THP-I cell line towards lactoferrin or the inflammatory or immune mediator MCP-I was tested in a chemotaxis or chemoattraction cell migration assay using the Chemicon ECM512 96-well kit with 175,000 cells per well after 2 hours of incubation. Lactoferrin alone was used to test its ability to function as a chemokine. Also fixed doses of lactoferrin were added to the increasing doses of MCP-I to measure their effect on the biological readout (cell migration). [0388] 175,000 cells in lOOul volume were loaded per well of a Chemicon ECM512 plate (5 um pore size) containing 96 wells. 150ul of serum free media containing the chemokine of choice was loaded in the appropriate feeder tray well. The migration chamber was inserted, the cells mixed cells and loaded. Plates were incubated at 37 2C for 2 hours. The migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 2C, periodically tilting the plate to help dislodge cells. 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer. FIG. IA shows that lactoferrin can attract as many THP-I cells as MCP-I. TLF = lactoferrin. FIG. IB shows that when combined with MCP-I at low doses (0.1 mg/ml), lactoferrin blocks THP-I cell migration towards MCP-I. LF = lactoferrin. FIG. 1C shows that when combined with MCP-I at high doses (1 mg/ml), lactoferrin enhances THP-I cell migration towards MCP-I.
Example 3 Chemoattractant activity of lactoferrin
[0389] The migration of different cells and cell lines are tested towards a source of lactoferrin as a chemokine in a chemotaxis or chemoattraction cell-migration assay such as the Chemicon ECM 96-well kit with 175,000 cells per well. The different cells are from human, mouse or other species and include T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
[0390] Jurkat, a human T cell line, and mouse blood cells were tested for chemoattraction to lactoferrin. Blood was collected using anticoagulants (heparinized tubes) and pooled. Volume was 3.5ml for WT (6 mice), 2.5ml for KO (5 mice). One volume of PBS was added. Lympholyte-Mammal (Cedarlane Labs) was gently layered under the diluted blood. Tubes were spun at 800g for 20 minutes and the top layer (serum) was aspirated to a few millimeters above the buffy coat. The buffy coat containing white blood cells (WBC) was collected; next the Lympholyte layer containing granulocytes was collected to just above the pellet containing the red blood cells and dead cells. Samples were brought to 15ml with serum free RPMI and spun again for 10 minutes. Pellets were brought up in 2ml Pharmlyse and incubated for 5 minutes at room temperature in the dark. Samples were diluted to 15ml with serum free RPMI and spun for 10 minutes. Pellets were brought up in 5ml media and counted. 175,000 cells in lOOul volume were loaded per well of a Chemicon ECM515 plate (3 urn pore size) containing 96 wells. 150ul of serum free media containing the chemokine of choice was loaded in the appropriate feeder tray well. The migration chamber was inserted, the cells mixed cells and loaded. Plates were incubated at 37 2C for 2 hours. The migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 2C, periodically tilting the plate to help dislodge cells. 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer , gating and counting separately the lymphocytes, granulocytes, macrophages and total cell count for each sample/fraction.
[0391] FIG. 2A shows that lactoferrin attracts Jurkat cells, a human T cell line, thus functioning as a chemokine for these cells. FIG. 2B shows that lactoferrin attracts mouse granulocytes, thus functioning as a chemokine for these cells. Granulocyes from IL-8 receptor^ knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of granulocytes. FIG. 2C shows that lactoferrin attracts mouse lymphocytes, thus functioning as a chemokine for these cells. Lymphocytes from IL-8 receptor knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of lymphocytes. FIG. 2D shows that lactoferrin attracts mouse macrophages, thus functioning as a chemokine for these cells. Macrophages from IL-8 receptor knock-out mice were attracted less efficiently, suggesting the IL-8 receptor is partly involved in mediating lactoferrin-induced chemotaxis of macrophages.
Example 4 Modulation of other chemokine's chemoattractant activity by lactoferrin
[0392] The migration of different cells and cell lines are tested towards a source of chemokine (increasing doses) plus different fixed doses of lactoferrin in a chemotaxis or chemoattraction cell-migration assay such as the Chemicon ECM512 96-well kit with 175,000 cells per well to test blockage or enhancement of cellular migration. The different cells are from human, mouse or other species and include T cell lines such as Jurkat, antigen presenting cells (APCs) such as dendritic cells, macrophages and B cells, natural killer cells, neutrophils, granulocytes, and peripheral blood mononuclear cells (PBMCs), splenocytes, lymph node cells, Peyer's Patches cells.
Example 5
Binding of lactoferrin to cells: flow cytometry
[0393] Cultured human intestinal HT-29 cells derived from colon carcinoma or normal human dermal fibroblasts (NHDF) from adult are exposed to different doses of lactoferrin, at 3-4 x 105 cells per dose, for 30 minutes at 4 0C in medium alone or with 2% FBS with or without 5% BSA. Cells are then washed 3 times, placed in staining buffer consisting of PBS with 0.5% (5 mg/ml) BSA and 5% heat-inactivated normal rabbit serum, incubated with biotinylated anti-lactoferrin antibody for 30 min, washed 3 times, incubated with streptavidin-TR (Texas red) in staining buffer, and binding analyzed by flow cytometry on a fluorescent activated cell sorter (FACS) system to obtain the percent cells staining positive and the mean fluorescence intensity (MFI).
[0394] FIG. 3 shows that lactoferrin binds to human intestinal and fibroblastic cells in a dose-dependent and saturating fashion in the presence of excess of other proteins, thus suggesting specific binding. FIG.3A: human intestinal HT-29 cells. FIG. 3B: human fibroblasts (NHDF cells) in the presence of 5% BSA.
Example 6
Binding of lactoferrin to cells: immunofluorescence
[0395] Cells were cultured in medium containing FBS for 2 hours at 370C or 1 hr at 40C with recombinant human lactoferrin in duplicate at different concentrations. Thirty minutes before the end of the incubation at 370C, the medium containing lactoferrin was changed to culture medium containing 10% FBS. Cells were washed 5 times with NaCl/Pi, and fixed with 4% paraformaldehyde in NaCl/Pi at 4°C for 30 minutes. Cells were washed with NaCl/Pi, permeabilized with 0.15% Triton-X-100 in NaCl/Pi for 2 minutes at 2O0C, washed again, and blocked with blocking buffer (0.15% Triton X-100, 1% BSA and 3% normal serum in PBS) for 30 minutes at 2O0C. The cells were then incubated with primary antibody (mouse monoclonal anti-human lactoferrin; Biodesign #H86024M; 1 :2000 dilution) for 1 hour, followed by biotinylated secondary antibody for 30 minutes, and streptavidin-FITC for 30 minutes. Secondary antibody alone was used as a control to determine specificity. The cells were counterstained with PI and cover-slipped with anti-fade mounting medium. Cells were examined under a fluorescent microscope and digital images taken from each chamber.
[0396] FIG. 4 shows specific binding of lactoferrin to human intestinal HT-29 cells, fibroblastic NHDF cells and hepatocytic HepG2 cells.
Example 7 Binding of lactoferrin to cells in vivo
[0397] An open, full-thickness wound was inflicted to normal mice (10 mm in diameter). Care was taken to avoid vessels during injury by elevating the redundant skin and using trans-illumination to avoid vessels. Lactoferrin solution at 8.5% was incubated on the fresh wound for five minutes, then washed five times with ice-cold saline. The entire wound with adjacent normal tissue was excised, fixed overnight in formalin, embedded in paraffin and sections from the wound margins prepared for confocal microscopy. The wall of the wound edge was stained with polyclonal rabbit anti-LF plus secondary goat anti-rabbit-FITC or stained with secondary-FITC only (x40 original magnification).
[0398] Fig. 5 shows specific binding of lactoferrin to dermal cells in the wall of an open wound (5A), which disappears when incubating without the specific antibody (only with the labeling antibody, 5B). Green: anti-LF staining.
Example 8
Binding of lactoferrin to cells in vivo
[0399] A full-thickness circular dermal punch wound (10 mm in diameter) was inflicted on the stomach of shaved, anesthetized mice. Care was taken to avoid vessels during injury by elevating the redundant skin and using trans-illumination to avoid vessels. The wounds were then filled with vehicle (PBS) or three different strengths of lactoferrin solution and incubated for five minutes. After washing 5 times with ice-cold saline, the entire wound with 5 mm of adjacent normal tissue was excised to the level of muscle and fat, fixed overnight in 10% neutral buffered formalin, embedded in paraffin, and 5-μm sections prepared from the wound margin. The sections were blocked with normal horse serum and stained with monoclonal mouse anti-lactoferrin antibody (Biodesign) at 1 :20,000 dilution followed by biotinylated horse anti-mouse secondary antibody and Streptavidin-FITC (Vector Laboratories) for detection. Nuclei were counterstained with PI, sections examined under a fluorescent microscope and digital images taken (original magnification x20). Sections without primary antibody served as a negative control.
[0400] FIG. 6 shows specific binding to dermal cells in an open wound only when lactoferrin is added to the wound and is absent when PBS is added. Binding increases with increasing doses of lactoferrin. Anti-lactoferrin antibodies are seen in green and nuclei in red. Arrows denote the wound margin; circles denote the dermal portion of the wound wall; (e) keratinocytic epithelium; (d) dermis; (hf) hair follicles; fm (fat and muscle). Original magnification: x20.
Example 9 Ability of lactoferrin to bind receptors for chemotactic, inflammatory or immune mediators
[0401] Specific binding in vitro of lactoferrin to different purified receptors assessed radioactively by the extent by which lactoferrin blocks the binding of a reference ligand to its particular cognate receptor. Recombinantly produced receptors were incubated alone to obtain the baseline 0% specific Tending or with an excess of their most common ligand (reference compound) radioactively labeled to obtain the baseline 100% specific binding (nonspecific binding level). Then, increasing doses of lactoferrin were added in the presence of the radio-ligand to determine the level of binding inhibition by comparing to the baseline values. The material was then precipitated and radioactivity counted. Equal or over 50% inhibition of specific binding by lactoferrin is considered significant. IC50 is the concentration causing 50% inhibition and is determined after a range of 9 concentrations of lactoferrin is tested. A related parameter, the inhibition constant (K1) is also determined, both giving an indication of the ability of lactoferrin to interfere with binding of the reference ligand for its receptor. Ki is calculated with the Cheng Prusoff equation: K,= IC5o/(l+ (L/Kd)) where L is the concentration of radioligand used in the assay and Kd is the affinity of the radioligand for the receptor.
[0402] FIG. 7 shows that lactoferrin binds specifically to receptors of chemotactic, inflammatory and immune mediators such as chemokines in a dose-dependent fashion as determined by the IC5Q and Ki. IL-8 beta receptor is involved in neutrophil attraction, CCR2 is the receptor for MCP-I and CCR3 is a receptor for MCP-2, MCP-3, MCP-3, MIP-Ia, MIP Ib, eotaxin 1-3, and RANTES. MCPs (macrophage chemotactic proteins) and MIPs (macrophage inflammatory proteins) are involved in wound healing and innate immune responses, Rantes in T cell activation and eotaxins in allergic/asthmatic responses.
Example 10 Ability of Iactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors in vivo in the setting of a wound condition [0403] BALB/c mice (n=4/group) received open, full-thickness wounds by punch biopsy and were treated daily with topically applied Iactoferrin (1%) or placebo gels. Mice were sacrificed on Day 3, wounds excised, homogenized in the presence of protease inhibitors and analyzed for cytokines using the Luminex system or ELISA kits.
[0404] FIG. 8 shows that Iactoferrin significantly induced chemokines MIP- lα and MIP-2 (mouse IL-8), and cytokines IL-6 and TNF-α in wounds in vivo. Significance was calculated versus placebo at each time point using a 2-tailed Student t test. * P < 0.05; ** P< 0.01.
Example 11 Ability of Iactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors: fibroblasts
[0405] Normal human dermal fibroblasts (NHDF) from adult (Cambrex) where grown in FGM-2 media with 2% FBS (Cambrex). Cells were exposed to different concentrations of recombinant human Iactoferrin. At different time points, media was collected, stored at —80 0C, and later analyzed for immune and inflammatory mediators such as cytokines, chemokines, and growth factors using the Luminex system. PMA was used as a positive control at a final concentration of 10 ng/ml dissolved in ethanol .
[0406] FIG. 9 shows that Iactoferrin (rhLF) induced the production of MCP-I (A), IL-8 (B), and IL-6 (C) in a time- and dose-dependent form. Example 12
Ability of lactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors: keratinocytes
[0407] Primary keratinocyte cultures (UCSD isolates from human neonatal foreskin epidermis) were grown to confluence and were incubated with placebo or lactoferrin at the concentrations shown. Supernatants were collected at the indicated time points and assayed by ELISA for MIP-3α. N=3 per group.
[0408] FIG. 10 shows that lactoferrin induces the chemokine MIP-3α from keratinocytes in a time and dose dependent fashion. * P < 0.05; ** P< 0.01
Example 13 Ability of lactoferrin to activate the biological activity of chemotactic, inflammatory or immune receptors: hepatocytes
[0409] Human hepatocytes, HepG2 cells, were grown in a 96-well format plate, employing conditions established previously for their optimum growth, and then tested for production of chemokine CCL20 (MIP-3α). Four (4) wells per group/conc./time point were used. Cells were exposed to various concentrations of placebo or test compounds: recombinant human lactoferrin from ^Agennix and native lactoferrin purified from human milk (Sigma). Supernatants (75 microL aliquots) were collected from cells at 2 h and 4 h after the administration of test compounds. At the end of the experiment (4 h sample collection), the cells were washed twice with the medium, then lysed using a Protease Inhibitor Cocktail medium and a homogenizer (e.g., using 20 strokes with a Dounce homogenizer). The cell homogenates was clarified by centrifugation at 1000xg for 20 minutes, supernatant filtered through a 0.22 μm filter, aliquoted into 100 μl aliquots, and stored at -80 0C until testing.
[0410] FIG. 11 shows that lactoferrin (rhLF) induces the chemokine CCL20 (MIP- 3α) in dose dependent manner at 2 h.
[0411] Using a 24- well format, HepG2 cells were grown to near confluence (with each well having about 1 MM of cells at confluence). The cells were incubated in vitro with the test compounds at different concentrations and times. At the end of each required period of incubation, supernatants were collected (in two aliquots of 250 μL) from each well and store frozen for cheraokine analysis. Cells were collected in RNAlater and stored for mRNA analysis by QC-PCR.
[0412] FIG. 12 shows the effect of various concentrations of lactoferrin at various time points on the protein production for MIP-3-α (A) and its mRNA message level (B). Induction by lactoferrin of these biomarkers in hepatocytes is both time and dose dependent.
[0413] Human hepatocytes (Hep-G2 cells) were exposed to various concentrations of lactoferrin or vehicle control for 6 hours. Supernatants were collected and analyzed using RayBiotech, Inc. Human Cytokine Antibody Array.
[0414] FIG. 13 shows the induction of chemotactic, inflammatory and immune mediators in human hepatocytes as the % change with lactoferrin at 40 mg/niL relative to vehicle treatment. Several inflammatory and immune mediators were increased significantly with lactoferrin treatment, including several chemokines such as MIP-3α, IL-8, TECK, I-TAC, Lymphotactin, PARC, MIP- lδ, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, and Eotaxin-3.
Example 14 Lactoferrin screen to find inhibitors of chemotactic, inflammatory or immune mediators
[0415] Same method of examples 9-12 is followed except that different doses of potential inhibitors are added concurrently with lactoferrin to test their ability to inhibit the production of chemotactic, inflammatory or immune mediators such as chemokines or other chemotactic modulators, growth factors, and cytokines, compared to a group without added inhibitor.
Example 15
Modulation of immune cell numbers by oral lactoferrin
[0416] Groups of six BALB/c mice were dosed once daily for three consecutive days with oral lactoferrin (2.9 g/m2) or placebo. Mice were injected subcutaneously with a mammary cancer cell line (TUBO) on the third day. On the fourth day, mice were sacrificed and FACS analysis conducted on a single-cell suspension of Peyer's patches isolated from the proximal small intestine. The number of cells in the Peyer's patches increases with lactoferrin relative to placebo.
[0417] FIG. 14 shows that oral lactoferrin (LF) increases the number of CD4+ T cells, CD8+ T cells, and Natural killer T cells (NKT) in a mouse immune organ (Peyer's patches), suggesting that lactoferrin-induced chemotaxis played a role in the process. * p<0.01.
Example 16 Modulation of immune cell migration by lactoferrin chemoattraction in vivo
[0418] Wild type (WT) control mice or IL-8 receptor (IL-8R) knock-out (KO) mice were administered lactoferrin orally: From day 3 to 12 mice (KO and ctrls) were dosed with lactoferrin (LF) at 1000 mg/kg for a total of 8 doses once per day each. On day 13 dosing was terminated and on day 18 mice were sacrificed and the Peyer's Patches were dissected. The Peyer's Patches were then placed in 24-well tissue culture plate in 250 ul RPMI with 10% FBS, 1% PS. Cells were ground up in plate, using the top of a sterile syringe plunger. 2.0 ml RPMI (with FBS plus penicillin and streptomycin) were added to each well, for a 2.25 ml total volume. In a 96-well plate, 10 ul of Trypan blue were mixed with 50 ul cell suspension, placing 10 ul of the mix into a hemacytometer followed by counting twice diagonally on middle square. Count was multiplied by 2^5 to yield number of cells x 104/ml.
[0419] FIG. 15 shows that the absence of the IL-8R impaired significantly (*p=0.0396) the increase in the total number of cells in a mouse immune organ (Peyer's Patches) induced by lactoferrin, suggesting that lactoferrin chemoattraction is mediated at least in part by IL- 8R in vivo.
Example 17
Modulation of immune cells by lactoferrin chemoattraction in situ [0420] Lactoferrin is directly injected into the Peyer's patch or other types of lymph nodes in mice. At different time points the node is extracted, the number of node cells determined, the percent expression of immune cells determined, and both measurements are compared with nodes injected with vehicle placebo. An anti-lactoferrin antibody is injected together with lactoferrin to neutralize this effect and confirm the specificity of lactoferrin' s chemotactic activity. Example 18 Lactoferrin binding to cells of immune organs with concomitant induction of maturation and/or activation
[0421] Peyer's patches, mesenteric lymph nodes, splenocytes, and other immune organs are incubated with or without lactoferrin and then analyzed for lactoferrin binding and immune cell markers using FACS analysis.
[0422] FIG. 16 shows binding of lactoferrin after 24 hours at 37 2C to all tested immune cells in mouse splenocytes with concomitant increase in surface markers for natural killer (NK) cells [DX5], B cells [CD19], and dendritic cells [CD205], MFI = mean fluorescence intensity. FITC = green dye. PE = red dye.
Example 19 Lactoferrin Binds Dermal Fibroblasts In Vitro
[0423] FIG.17 Left Panel: Dermal fibroblasts were incubated with or without Lactoferrin in the presence of 5% bovine serum albumin for 30 min at 37C, stained with a biotinylated rabbit anti-lactoferrin antibody followed by streptavidin-TR, and analyzed using a fluorescent activated cell sorter (FACS) machine.
[0424] FIG.17 Right Panel: Fibroblasts were cultured in 2% FBS medium for 2 hours at 370C with Lactoferrin, fixed, permeabilized and blocked. The cells were then incubated with a mouse monoclonal anti-human lactoferrin followed by biotinylated secondary antibody and strepavidin-FITC. The cells were counter stained with PI, coverslipped with anti-fade mounting medium, and analyzed by fluorescent microscopy.
Example 20 Lactoferrin Induces the Movement of Fibroblasts
[0425] The migration of normal human dermal fibroblasts from adult towards lactoferrin was tested in a chemotaxis or chemoattraction cell migration assay using the Chemicon ECM512 96-well kit with 175,000 cells per well after 2 hours of incubation. [0426] 175,000 cells in 1 OOul volume were loaded per well of a Chemicon ECM512 (8 um pore size) plate containing 96 wells. 150ul of serum free media containing the chemokine of choice was loaded in the appropriate feeder tray well. The migration chamber was inserted, the cells mixed cells and loaded. Plates were incubated at 37 oC for 2 hours. The migration chamber was removed, discarding the content of wells, and the chamber placed in a new feeder tray with 150ul pre-warmed Cell Detachment Solution per well followed by incubation for 30 minutes at 37 oC, periodically tilting the plate to help dislodge cells. 75ul of cells from the lower tray media were then combined with 75ul from the corresponding well from the Cell Detachment Solution tray, mixing the cells thoroughly before harvesting. Cells were counted using a flow cytometer.
[0427] FIG. 18 shows that lactoferrin attracts normal human dermal fibroblasts from adult, thus functioning as a chemokine for these cells. Y axis is the mean chemotactic index defined as the mean number of migrated cells divided by the mean number of cells migrated with medium only (zero lactoferrin). X axis is lactoferrin concentration [micromolar].
Example 21 Orally administered lactoferrin localizes to Peyer's patches and binds key immune cells.
[0428] Lactoferrin binding to cells from the Peyer's patches using flow cytometry. Balb/C mice received oral lactoferrin (12.5 mmoles/kg) or vehicle (PBS). Animals were sacrificed at the indicated times and cells from the small intestinal Peyer's patches isolated. Extent of lactoferrin binding was determined by FACS analysis using different gates based on side and forward scatter properties to assess different cell populations. The experiment was performed twice with similar results. Positive control: Peyer's patches cells incubated in vitro with 1 uM lactoferrin. Negative control: untreated Peyer's patches cells (PBS or 0 TLF). Lactoferrin was also quantified in Peyer's patches by ELISA.
[0429] FIG.19A shows lactoferrin binding to cells from the Peyer's patches using flow cytometry, with a strong signal peaking early between 1 and 15 minutes after oral administration. Forward and side scatter properties of the cells suggested that granulocytes and macrophages are mainly responsible for the binding with lymphocytes mediating a low level of binding as well. FIG 19. B shows quantification of cell lysates by ELISA indicating substantial amounts of lactoferrin in Peyer's patches at 1-15 minutes following oral administration. Both approacnes suggest a range between 0.5-1 uM lactoferrin at the peak, following by degradation at later time points.
Example 22 Oral lactoferrin induces migration of dendritic cells to Peyer's patches
[0430] Lactoferrin was tested for its ability to induce the migration of dendritic cells into the Peyer's patches following oral administration. Mouse dendritic cells (DCs) were generated by culturing bone marrow cells in IL-4 and GM-CSF. After 6 days, a population ranging between 60%-70% CDl lc+ DCs was obtained. Cells were labeled with PKH2 (Sigma) and 106 CDl lc+ cells were injected into mice. Lactoferrin (1000 mg/kg) was administered by oral gavage starting either the day of the DC injection or 2 days prior to injection and continued for a further two days before animals were sacrificed. Peyer's Patches (5/animal) and spleens were collected and analyzed for labeled cells by flow cytometry. Cells from two or three animals within the same group were pooled to obtain enough cells for analysis.
[0431] FIG.20A shows that oral lactoferrin induces the migration of labeled dendritic cells into the Peyer's patches compared to untreated mice. Pre-treating the animals with lactoferrin [LF] for two days before the injection of labeled dendritic cells resulted in an enhancement of attraction Jo this organ. Accumulation in Peyer's patches appeared to be specific to this GALT tissue since such a response was not observed in spleen. In fact, compared to Peyer's patches, in the spleen there was an inverse trend of decreased dendritic cell accumulation with increased lactoferrin treatment, suggesting lactoferrin could be deviating the antigen presenting cells from a default response where a proportion of circulating dendritic cells migrate to the spleen. N=O per group. Data shown are mean + standard error. * P < 0.05 and ** P < 0.01 vs. Untreated group [t test, two tail]. FIG.20B shows that the induction of DC migration by oral lactoferrin disappears in CD 14 knockout mice indicating the CD 14 receptor is a potential mediator of this effect. N=4 for wild type [WT] treated group and N=2 for the remaining groups. * P < 0.05 [t test, one-tail].
REFERENCES CITED
[0432] All patents and publications mentioned in the specifications are indicative of the levels of those skilled in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference. U.S. Patent No. 4,554,101
U.S. Patent No. 5,220,007
U.S. Patent No. 5,284,760
U.S. Patent No. 5,354,670
U.S. Patent No. 5,366,878
U.S. Patent No. 5,389,514
U.S. Patent No. 5,571,691
U.S. Patent No. 5,571,697
U.S. Patent No. 5,571,896
U.S. Patent No. 5,629,001
U.S. Patent No. 5,635,377
U.S. Patent No. 5,789,166
U.S. Patent No. 5,919,913
U.S. Patent No. 6,066,469
U.S. Patent No. 6,080,559
U.S. Patent No. 6,100,054
U.S. Patent No. 6,277,817
U.S. Patent No. 6,228,614
U.S. Patent No. 6,333,311
U.S. Patent No. 6,455,687
Actor et al., Int Immunopharmacol 2002; 2:475-486.
Anderson et al., J MoI Biol. 1989; 209:711-734.
Bellamy et al., Med Microbiol Immunol. 1993; 182:97-105.
Bezault et al., Cancer Res. 1994; 54:2310-2312..
Cumberbatch et al., Immunology. 2000; 100:21-28.
Dhennin-Duthille et al., J Cell Biochem. 2000; 79:583-593.
Dial et al., J Pharm Pharmacol. 2000; 52:1541-1546.
Edde et al., Am. J. Physiol. Gastrointest. Liver Physiol. 2001 ; 281 :G1140-G1150
Edmonds et al., Metab Res Rev. 2000; 16 (Suppl 1):S51-S54.
Elias et al., J Clin Invest. 2003; 111 :291-297.
Elrod et al., Am J Respir Crit Care Med. 1997; 156:375-381.
Fernberg et al., Med Oncol. 2001; 8:231-235.
Gahr et al., J Leukocyte Biol. 1991 ;49: 427-433. Groves et al., Adv Skin Wound Care 2000; 13(3 Pt l):107-l 12.
Guillen et al., J Immunol. 2002; 168:3950-3957.
Headon, In: Lactoferrin: Structure, Function and Applications. Elsevier Science, B.V.,
Amsterdam, 2000; 415-427. Horowitz et al., J Immunol. 1984; 132:2370-2374. Ikeda et al., Virus Res. 2000; 66:51-63.
Inoue et al., Bone Marrow Transplantation 2001 : 28:1091-1092. Jaschke et al., Int J Dermatol. 1999; 38:380-386. Kampfer et al., J Invest Dermatol. 1999; 113:369-374. Kampfer et al., Eur Cytokine Netw. 2000; 11 :626-633. Kampfer et al., MoI Med. 2000; 6:1016-1027. Kimber et al., In: Lactoferrin: Structure, Function and Applications. Elsevier Science B.V.
2000;87-93.
Kimber et al., Biochem Cell Biol. 2002; 80:103-107. Kuhara et al., Nutrition and Cancer 2000; 38:192-199. Kuhn et al., In Basic Fibroblast Growth Factor in a Carboxymethylcellulose Vehicle Reverses the Bacterial Retardation of Wound Contraction Wounds 2001; 13:73-80. Lee et al., Infect Immun. 1998; 66:1421-1426. Lu et al., Int Jϋematol. 1991; 54:117-124. Mandracchia et al., Clin Pod Med Surg. 2001; 18:189-209. Mann et al., J Invest Dermatol. 2001; 117:1382-1390. Mantey et al., Commun Dis Public Health 2000; 3:288-2890. Marchetti et al., Biometals. 1998; 11:89-94. Metz-Boutigue et al., Eur J Biochem. 1984; 145:659-676. Muller et al., Mycoses. 1999; 42 Suppl 2:77-82. Norrby et al., Int J Cancer 2001; 91 :236-240. Okamura et al., Nature 1995; 378:88-91. Romagnani S. Ann Allergy Asthma Immunol. 2000; 85:9-18. Semba et al., AIDS 1998; 12:331-332. Shau et al., J Leukocyte Biol. 1992; 51 :343-349. Shimizu et al., Arch Virol. 1996; 141 :1875-1889. Singh et al., Immunol Res. 1999; 20:147-161 Spik et al., Adv Exp Med Biol. 1994; 357:13-19. Superti et al., Med Microbiol Immunol (Berl). 1997; 186:83-91.
Suzuki et al., Biochem Cell Biol. 2002;80(l):75-80.
Swart et al., Adv Exp Med Biol. 1998; 443:205-213.
Takayama et al., FEBS Lett. 2001; 508:111-116.
Takayama et al., Biochem Biophys Res Commun. 2002; 299:813-817.
Takayama et al., J. Biol Chem 2003; 278:22112-22118.
Ushio et al., J Immunol. 1996; 156:4274-4279.
Wakabayashi et al., J Med Microbiol. 2002; 51 :844-8450.
Wakabayashi et al., Curr Pharm Des. 2003; 9:1277-1287.
Ward et al., Biotechnol. Genet Eng Rev. 1997; 14:303-319.
Ward et al., Biotechnology 1995; 13:498-503.
Ward et al., Lactoferrin and Host Defense Biochem Cell Biol. 2002; 80:95-102.
Zhang et al., Gene Ther. 2001; 8:1333-1342.
[0433] Although the present invention and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined by the appended claims. Moreover, the scope of the present application is not intended to be limited to the particular, embodiments of-the process, machine, manufacture, composition of matter, means, methods and steps described in the specification. As one of ordinary skill in the art will readily appreciate from the disclosure of the present invention, processes, machines, manufacture, compositions of matter, means, methods, or steps, presently existing or later to be developed that perform substantially the same function or achieve substantially the same result as the corresponding embodiments described herein may be utilized according to the present invention. Accordingly, the appended claims are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or steps.

Claims

1. A method of treating a disease or condition in a subject comprising the step of administering to the subject a chemotactic modulator composition in an amount sufficient to modulate chemotactic activity in said subject.
2. The method of claim 1, wherein the disease or condition is selected from the group consisting of a hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
3. The method of claim 1, wherein chemotactic modulator is selected from the group consisting of lactoferrin, chemokine, chemokine receptor, inflammatory or immune mediator, viral anti-chemoldne, a mixture of casein and autologous serum, chemotactic C5 fragment, formyl-1-methionyl-l-leucine-l-phenylalanine (f-Met-Leu-Phe), bacterial factor derived from Escherichia coli, the simple peptide formylmethionylalanine, and activated human complement, carbamylcholine, phenylephrine, and cyclic guanosine 5'- monophosphate, nucleosides and nucleotides and prostaglandins
4. A method of treating a disease or condition in a subject comprising the step of administering to a subject a lactoferrin composition in an amount sufficient to modulate chemotactic activity in said subject.
5. The method of claim 4, wherein modulation refers to increase.
6. The method of claim 4, wherein modulation refers to decrease.
7. The method of claim 4, wherein chemotactic activity refers to attraction of cells by lactoferrin's ability to act as a chemokine.
8. The method of claim 4, wherein chemotactic activity refers to an activity that influences the movement or migration of cells either through the induction or inhibition of other chemotactic modulators.
9. The method of claim 4, wherein said lactoferrin composition is dispersed in a pharmaceutically acceptable carrier.
10. The method of claim 4, wherein said lactoferrin is mammalian lactoferrin.
11. The method of claim 4, wherein said lactoferrin is human or bovine.
12. The method of claim 4, wherein said lactoferrin is recombinant lactoferrin.
13. The method of claim 4, wherein said lactoferrin is produced by purification from natural sources or artificially.
14. The method of claim 4, wherein said lactoferrin composition comprises an amino acid sequence selected from the group consisting of SEQ.ID.NO.l, SEQ.ID.NO.2, SEQ.ID.NO.3, SEQ.ID.NO.4, SEQ.ID.NO.5, SEQ.ID.NO.6, SEQ.ID.NO.7, SEQ.ID.NO.8, SEQ.ID.NO.9, SEQ.ΪD.NO.10, and SEQ.ID.NO.i l.
15. The method of claim 4, wherein said lactoferrin is administered topically, orally or parenterally.
16. The method of claim 4, wherein the amount of the composition that is administered is about lμg to about lOOg per day.
17. The method of claim 4, wherein said lactoferrin composition is a topical gel, a solution, or a tablet containing a concentration of about 0.01% to about 20% of lactoferrin.
18. The method of claim 17, wherein said topical gel is composed from the polymers selected from the group of carbopol, carboxymethyl-cellulose, and pluronic polymers.
19. The method of claim 4, wherein the disease or condition are selected from the group consisting of hyperproliferative disease, respiratory disorder, cardiovascular disease, neurological condition, autoimmune disorder, inflammatory disorder, gastrointestinal disorder, endocrine and/or metabolism disorder, hematological disorder, ocular disorder, integument disorder, pain, and fibrotic disease.
20. The method of claim 19, wherein the skin disease is selected from the group consisting of psoriasis, scleroderma, systemic lupus erythematosus, atopic dermatitis, epidermolysis bullosa, acute wounds, chronic wounds, and burns.
21. The method of claim 19, wherein the gastrointestinal disorder is Crohn's disease, colitis, necrotizing entercolitis, endometriosis, irritable bowel syndrome, pancreatitis, periodontal disease, and ulcerative colitis.
22. The method of claim 19, wherein the fibrotic disease is selected from the group consisting of diabetic nephropathy, retinopathy, and other forms of renal fibrosis, as well as pulmonary fibrosis, organ transplant rejection, liver fibrosis, and atherosclerosis.
23. The method of claim 19, wherein the inflammatory condition is of an infectious origin or a non-infectious origin.
24. The method of claim 23, wherein the infectious inflammatory disease is casued by a pathogen selected from the group consisting of meningococci, Salmonella typhi, Dengue virus, Plasmodia, Pneumocystis carinii, Yersinia enterocolitica pathogens, and enterovirus, fungi.
25. The method of claim 23, wherein non-infectious inflammatory disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, organ-specific inflammatory diseases, glomerular disease, Sjogrens disease, Hashimoto's thyroiditis, chronic skin diseases, chronic obstructive pulmonary disease, periodic disease, wound healing and posttraumatic dystrophia, non-bacterial sepsis, multi-trauma and multi-organ dysfunction syndrome.
26. The method of claim 19, wherein the wound condition is selected from the group consisting of chronic wound, acute wound, skin wound, internal wound, gastrointestinal wound, oral wound, ophthalmic wound, surgical wound, incisional wound, excisional wound, diabetic ulcer, venous stasis ulcer, decubitus or pressure ulcer, chemical wound, burn wound, full-thickness skin wound and partial-thickness skin wound.
27. The method of claim 19, wherein the autoimmune disease is selected from the group consisting of Active Chronic Hepatitis, Addison's Disease, Anti-phospholipid Syndrome, Atopic Allergy, Atrophic Gastritis, Achlorhydra Autoimmune, Celiac Disease, Crohn's Disease, Cushings Syndrome, Dermatomyositis, (Type I) Diabetes, Discoid Lupus, Erythematosis, Goodpasture's Syndrome, Grave's Disease, Hashimoto's Thyroiditis, Idiopathic Adrenal Atrophy, Idiopathic Thrombocytopenia, Insulin-dependent Diabetes, Lambert-Eaton Syndrome, Lupoid Hepatitis, Lymphopenia, Mixed Connective Tissue Disease, Multiple Sclerosis, Pemphigoid, Pemphigus Vulgaris, Pernicious Anema, Phacogenic Uveitis, Polyarteritis Nodosa, Polyglandular Auto Syndromes, Primary Biliary Cirrhosis, Primary Sclerosing Cholangitis, Psoriasis, Raynauds, Reiter's Syndrome, Relapsing Polychondritis, Rheumatoid Arthritis, Schmidt's Syndrome, Scleroderma-CREST, Sjogren's Syndrome, Sympathetic Ophthalmia, Systemic Lupus Erythematosis, Takayasu's Arteritis, Temporal Arteritis, Thyrotoxicosis, Type B Insulin Resistance, Ulcerative Colitis, Wegener's Granulomatosis, graft versus host disease (GVHD), organ transplant rejection, autoimmune hepatitis, autoimmune cholangitis, irritable bowel syndrome (IBS), chronic granulomatous disease, ankylosing spondylitis, scleroderma, polymyositis, (dermato)myositis, and systemic vasculitis.
28. The method of claim 19, wherein the hyperproliferative disease is further defined as cancer.
29. The method of claim 28, wherein the cancer comprises a neoplasm.
30. The method of claim 29, wherein the neoplasm is selected from the group consisting of melanoma, non-small cell lung, small-cell lung, lung hepatocarcinoma, retinoblastoma, astrocytoma, gliobastoma, leukemia, neuroblastoma, squamous cell, head, neck, gum, tongue, breast, pancreatic, prostate, renal, bone, testicular, ovarian, mesothelioma, sarcoma, cervical, gastrointestinal, lymphoma, brain, colon, and bladder.
31. The method of claim 29, wherein the neoplasm is a hematopoietic neoplasm.
32. The method of claim 30, wherein the hematopoietic neoplasm is selected from the group consisting of acute myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, and chronic lymphocytic leukemia.
33. The method of claim 19, wherein the hyperproliferative disease is selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, osteoarthritis, leiomyomas, adenomas, lipomas, hemangiomas, fibromas, vascular occlusion, restenosis, atherosclerosis, pre-neoplastic lesions, carcinoma in situ, oral hairy leukoplakia, and psoriasis.
34. The method of claim 19, wherein the endocrine and/or metabolism disorder is diabetes mellitus, thyroid disorder, or fragile bone disease.
35. The method of claim 33, wherein the fragile bone disease is selected from the group consisting of osetoporosis, osteitis deformans, osteogenesis imperfecta, osteopetrosis, Rickets, and osteomalacia.
36. The method of claim 19, wherein the pain is acute or chronic pain.
37. The method of claim 19, wherein the cardiovascular disease is congestive heart failure, hypertension, cardiomyopathy, myocarditis, atherosclerosis, chronic venous disease, or heart arrhythmia.
38. The method of claim 19, wherein the hematological disorder is anaemia, sickle cell anemia or cachexia.
39. The method of claim 19, wherein the ocular disorder is conjunctivitis, dry eye disease, glaucoma, allergic eye disease, uveitis or ocular infection.
40. The method of claim 19, wherein the respiratory disorder is atopic asthma, non-atopic asthma, emphysema, bronchitis, chronic obstructive pulmonary disease, sinusitis, allergic rhinitis, fibrotic lung disease, COPD, ARDS, pulmonary vascular disease/pulmonary hypertension, Cor Pulmonale, or cystic fibrosis.
41. The method of claim 4, further comprising administering a subject a lactoferrin composition in combination with other appropriate therapy.
42. A method of treating a disease or condition in a subject by administering to the subject a sufficient amount of a lactoferrin composition, wherein the lactoferrin composition binds to receptors of chemotactic, immune, or inflammatory mediators.
43. The method of claim 42, wherein the chemotactic, immune or inflammatory mediators are chemokines and other chemotactic modulators, cytokines, or growth factors.
44. The method of claim 42, wherein disease or condition are selected from the group consisting of skin disease, fibrotic disease, chronic inflammatory disease, wound condition, inflammatory condition, autoimmune disease, hyperproliferative disease, fragile bone disease, pain and diabetes mellitus.
45. The method of claim 42, wherein lactoferrin acts directly as a chemokine to attract cells.
46. The method of claim 42, wherein the binding of lactoferrin to the receptors inhibits the chemotactic, immune or inflammatory mediators.
47. The method of claim 42, wherein the binding of lactoferrin to the receptors stimulates the chemotactic, immune or inflammatory mediators.
48. The method of claim 42, wherein the chemokines are MIP-lalρha, MlP-lβ, IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha, TECK, I-TAC, Lymphotactin, PARC, MIP-I delta, C-TACK, TARC, HCC-4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO-α, GRO-β, GRO-γ, SCF, or MEC/CCL28.
49. The method of claim 42, wherein the receptors are IL-8 alpha receptor, IL-8 beta receptor, neuropeptide Yl receptor, PACAP receptor, K+ channel receptor, CCR2, CCR3, CCR6, CXCR4, EGFR, TLR-2, TLR-3, TLR-4, TLR4/MD, Mannose receptor, CD 14, or TRAIL R4.
50. A method of stimulating a cultured cell to induce chemotactic, inflammatory and/or immune mediators comprising the step of adding a lactoferrin composition to the cell thereby stimulating MIP-I alpha, MIP- lβ, IL-8, MIP-2, MCP-I, MCP-2, MCP-3 MCP-4, MIP-3alpha, TECK, I-TAC, Lymphotactin, PARC, MIP-I delta, C-TACK, TARC, HCC- 4, RANTES, Eotaxin, MCP-2, MCP-3, MCP-4, CCL-28, Eotaxin-2, Eotaxin-3, GRO-α, GRO-β, GRO-γ, SCF, or MEC/CCL28.
PCT/US2006/032803 2005-08-19 2006-08-21 Use of lactoferrin as a chemokine and a chemotactic modulator WO2007022537A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US70973505P 2005-08-19 2005-08-19
US60/709,735 2005-08-19

Publications (2)

Publication Number Publication Date
WO2007022537A2 true WO2007022537A2 (en) 2007-02-22
WO2007022537A3 WO2007022537A3 (en) 2007-10-11

Family

ID=37758510

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/032803 WO2007022537A2 (en) 2005-08-19 2006-08-21 Use of lactoferrin as a chemokine and a chemotactic modulator

Country Status (1)

Country Link
WO (1) WO2007022537A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010042788A1 (en) * 2008-10-10 2010-04-15 Celtaxsys, Inc. Method of inducing negative chemotaxis
WO2012077076A1 (en) * 2010-12-08 2012-06-14 Holt Patrick G Treating or preventing sensitivity to milk allergens
US20120172288A1 (en) * 2010-12-29 2012-07-05 Anja Wittke Use of nutritional compositions including lactoferrin in supporting resistance to diseases and conditions
EP2560677A1 (en) * 2010-04-23 2013-02-27 Probiotec Limited Cold treatment
EP2560679A1 (en) * 2010-04-23 2013-02-27 Probiotec Limited Eczema treatment
EP2658396A1 (en) * 2010-12-29 2013-11-06 MJN U.S. Holdings LLC Use of nutritional compositions including lactoferrin in stimulating immune cells
WO2014029924A1 (en) * 2012-08-23 2014-02-27 Allain Henri Novel uses of lactoferrin and pharmaceutical and nutraceutical compositions
AU2011269657B2 (en) * 2010-06-24 2015-07-30 Murray Goulburn Co-Operative Co. Limited Method of treatment
CN105101989A (en) * 2013-04-09 2015-11-25 国立大学法人东京大学 Inhibitor of extracellular trap formation in leukocytes
RU2575776C2 (en) * 2010-12-29 2016-02-20 ЭмДжейЭн Ю. Эс. Холдингс ЛЛК Application of lactoferrin-containing nutritional compositions for stimulation of immune cells
EP3205347A4 (en) * 2014-10-08 2018-06-06 Keio University White blood cell extracellular trap formation inhibitor
WO2019171236A1 (en) 2018-03-09 2019-09-12 Frimline Private Limited A pharmaceutical composition for anaemia
WO2020018440A1 (en) * 2018-07-17 2020-01-23 Cellastra Inc. Methods for regulating endogenous production of lactoferrin and sub-peptides thereof
EP3815707A4 (en) * 2018-06-26 2022-04-06 S&K Biopharma, Inc. Glycosaminoglycan inhibitor and promoter
EP4137511A4 (en) * 2020-04-16 2024-01-10 Dermopartners, S.L. New composition for use to treat and prevent infections by covid-19 and other coronaviruses

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040009895A1 (en) * 2002-05-10 2004-01-15 Atul Varadhachary Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US20050004006A1 (en) * 2003-05-14 2005-01-06 Jose Engelmayer Lactoferrin in the treatment of diabetes mellitus

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040009895A1 (en) * 2002-05-10 2004-01-15 Atul Varadhachary Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US20050004006A1 (en) * 2003-05-14 2005-01-06 Jose Engelmayer Lactoferrin in the treatment of diabetes mellitus

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8377914B2 (en) 2008-10-10 2013-02-19 Celtaxsys, Inc. Method of inducing negative chemotaxis
WO2010042788A1 (en) * 2008-10-10 2010-04-15 Celtaxsys, Inc. Method of inducing negative chemotaxis
EP3210618A1 (en) * 2010-04-23 2017-08-30 Probiotec Limited Composition comprising lactoferrin and immunoglobulin for the treatment of eczema
EP2560677A1 (en) * 2010-04-23 2013-02-27 Probiotec Limited Cold treatment
EP2560679A1 (en) * 2010-04-23 2013-02-27 Probiotec Limited Eczema treatment
EP2560677A4 (en) * 2010-04-23 2013-09-11 Probiotec Ltd Cold treatment
EP2560679A4 (en) * 2010-04-23 2013-09-18 Probiotec Ltd Eczema treatment
AU2011269657B2 (en) * 2010-06-24 2015-07-30 Murray Goulburn Co-Operative Co. Limited Method of treatment
WO2012077076A1 (en) * 2010-12-08 2012-06-14 Holt Patrick G Treating or preventing sensitivity to milk allergens
EP3205350A1 (en) * 2010-12-29 2017-08-16 MJN U.S. Holdings, LLC Infant formulas including lactoferrin for stimulating immune cells
US20120172288A1 (en) * 2010-12-29 2012-07-05 Anja Wittke Use of nutritional compositions including lactoferrin in supporting resistance to diseases and conditions
EP2658396A1 (en) * 2010-12-29 2013-11-06 MJN U.S. Holdings LLC Use of nutritional compositions including lactoferrin in stimulating immune cells
RU2575776C2 (en) * 2010-12-29 2016-02-20 ЭмДжейЭн Ю. Эс. Холдингс ЛЛК Application of lactoferrin-containing nutritional compositions for stimulation of immune cells
WO2014029924A1 (en) * 2012-08-23 2014-02-27 Allain Henri Novel uses of lactoferrin and pharmaceutical and nutraceutical compositions
FR2994657A1 (en) * 2012-08-23 2014-02-28 Henri Allain NEW APPLICATIONS AND USES OF LACTOFERRIN
CN105101989A (en) * 2013-04-09 2015-11-25 国立大学法人东京大学 Inhibitor of extracellular trap formation in leukocytes
EP3017824A4 (en) * 2013-04-09 2016-10-05 Univ Tokyo Inhibitor of extracellular trap formation in leukocytes
EP3205347A4 (en) * 2014-10-08 2018-06-06 Keio University White blood cell extracellular trap formation inhibitor
WO2019171236A1 (en) 2018-03-09 2019-09-12 Frimline Private Limited A pharmaceutical composition for anaemia
EP3815707A4 (en) * 2018-06-26 2022-04-06 S&K Biopharma, Inc. Glycosaminoglycan inhibitor and promoter
WO2020018440A1 (en) * 2018-07-17 2020-01-23 Cellastra Inc. Methods for regulating endogenous production of lactoferrin and sub-peptides thereof
US11891429B2 (en) 2018-07-17 2024-02-06 Cellastra Inc. Methods for regulating endogenous production of lactoferrin and sub-peptides thereof
EP4137511A4 (en) * 2020-04-16 2024-01-10 Dermopartners, S.L. New composition for use to treat and prevent infections by covid-19 and other coronaviruses

Also Published As

Publication number Publication date
WO2007022537A3 (en) 2007-10-11

Similar Documents

Publication Publication Date Title
US7420033B2 (en) Composition of lactoferrin related peptides and uses thereof
WO2007022537A2 (en) Use of lactoferrin as a chemokine and a chemotactic modulator
US11518782B2 (en) Compositions containing HC-HA/PTX3 complexes and methods of use thereof
JP5969457B2 (en) Compounds and their use
AU2014244744B2 (en) Pharmaceutical composition for inhibiting immune response through inducing differentiation into regulator T cells and promoting proliferation of regulator T cells
Liu et al. Hybrid biomaterial initiates refractory wound healing via inducing transiently heightened inflammatory responses
CA3004951A1 (en) Control of cellular redox levels
US7897180B2 (en) Enamel matrix protein composition for treatment of systemic inflammatory response
JP6158097B2 (en) Peptides for inhibiting inflammation
CN109328069A (en) Purposes of the IL-22 in treatment necrotizing enterocolitis
AU2017252344A1 (en) Diagnosis of immune_activation using CLEVER-1, TNF-alpha and HLA-DR binding agents
KR101620341B1 (en) Use of an apl peptide for the treatment of inflammatory bowel disease and type 1 diabetes
WO2024104197A1 (en) Peptide hydrogel, preparation method therefor and use thereof in treatment
KR101979932B1 (en) Peptide for Inhibiting Bone Resorption
WO2005018542A2 (en) Use of lactoferrin in prophylaxis against infection and/or inflammation in immunosuppressed subjects
WO2016056665A1 (en) White blood cell extracellular trap formation inhibitor
JP7000458B2 (en) Treatment of multiple sclerosis with long-acting glatiramer and adipose-derived stem cells
TWI726904B (en) Medical uses of fungal immunomodulatory proteins in treatment and prophylaxis of alveolar bone loss due to periodontal disease
JP2020527573A (en) Compositions and Methods for Treating or Ameliorating Scleroderma-Related Angiopathy
KR102372552B1 (en) Composition for preventing or treating immune disease comprising Alphalipoic acid and metformin
TWI373338B (en) Pharmaceutical composition containing transcription factor decoys and their preparation method and applications
WO2021226510A2 (en) Akt3 modulators and methods of use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06802098

Country of ref document: EP

Kind code of ref document: A2