US20040082504A1 - Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases - Google Patents

Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases Download PDF

Info

Publication number
US20040082504A1
US20040082504A1 US10/435,319 US43531903A US2004082504A1 US 20040082504 A1 US20040082504 A1 US 20040082504A1 US 43531903 A US43531903 A US 43531903A US 2004082504 A1 US2004082504 A1 US 2004082504A1
Authority
US
United States
Prior art keywords
lactoferrin
tumor
rhlf
cells
administered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/435,319
Other languages
English (en)
Inventor
Atul Varadhachary
Rick Barsky
Karel Petrak
Bert O'Malley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agennix Inc
Original Assignee
Agennix Inc
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 Inc filed Critical Agennix Inc
Priority to US10/435,319 priority Critical patent/US20040082504A1/en
Assigned to AGENNIX INCORPORATED reassignment AGENNIX INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BARSKY, RICK, PETRAK, KAREL, VARADHACHARY, ATUL
Publication of US20040082504A1 publication Critical patent/US20040082504A1/en
Assigned to GPC BIOTECH AG reassignment GPC BIOTECH AG SECURITY AGREEMENT Assignors: AGENNIX INCORPORATED
Priority to US12/561,429 priority patent/US20100137208A1/en
Assigned to AGENNIX INC. reassignment AGENNIX INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: O'MALLEY, BERT W., JR
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/17Amino acids, peptides or proteins
    • A23L33/19Dairy proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs

Definitions

  • the present invention relates to methods of treating a hyperproliferative disease by administering a composition of lactoferrin alone or in combination with standard anti-cancer therapies.
  • the lactoferrin composition may be administered orally, intravenously, intratumorally, or topically.
  • Radiotherapy chemotherapy, biotherapy and immunotherapy are alternatives to surgical treatment of cancer (Mayer, 1998; Ohara, 1998; Ho et al., 1998).
  • the disadvantage of many of the alternative therapies are the side effects, which can include myelosuppression, skin irritation, difficulty swallowing, dry mouth, nausea, diarrhea, hair loss, weight loss, and loss of energy (Curran, 1998; Brizel, 1998).
  • Lactoferrin is a single chain metal binding glycoprotein. Many cells types, such as 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 lymphokine-activated killer
  • bovine lactoferrin (bLF) was used as a prophylaxis for tumor formation and/or established tumors.
  • the present invention is the first to use lactoferrin as a treatment, not a prophylaxis, for established tumors.
  • the present invention is directed to a method for treating a hyperproliferative disease.
  • the method of treatment involves intratumoral administration of lactoferrin.
  • An embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of administering intratumorally to a subject a lactoferrin composition in an amount sufficient to provide an improvement in the hyperproliferative disease.
  • the amount of the lactoferrin composition that is administered is about 0.1 ⁇ g to about 10 ⁇ g per day.
  • the lactoferrin composition is dispersed in a pharmaceutically acceptable carrier. More particularly, the lactoferrin is mammalian lactoferrin, for example, human or bovine. In specific embodiments, the lactoferrin is recombinant lactoferrin.
  • the hyperproliferative disease is further defined as cancer, which comprises a neoplasm.
  • 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.
  • 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.
  • Another embodiment is a method of treating a hyperproliferative disease comprising the step of supplementing a systemic and/or local immune system in a subject by increasing the amount of lactoferrin in the vicinity of the hyperproliferative disease.
  • the lactoferrin is administered intratumorally.
  • the lactoferrin enhances, stimulates' and/or up-regulates interleukin-18 and Granulocyte Macrophage Colony Stimulating Factor (GM-CSF).
  • GM-CSF Granulocyte Macrophage Colony Stimulating Factor
  • interleukin-18 stimulates the production or activity of immune cells, for example T lymphocytes or natural killer cells and GM-CSF promotes the migration and maturation of immune cells including dendritic and other antigen presenting cells.
  • Another embodiment is a method of enhancing a local immune response in the vicinity of a tumor following the step of administering intratumorally to the subject a lactoferrin composition.
  • the lactoferrin composition stimulates interleukin-18 and/or GM-CSF in the site of injection, which stimulates the production or activity of immune cells, e.g., T lymphocytes or natural killer cells.
  • T lymphocytes are selected from the group consisting of CD4+, CD8+ and CD3+ cells.
  • the present invention is drawn to a method of treating a hyperproliferative disease comprising administering intratumorally to a subject a lactoferrin composition in combination with chemotherapy, biotherapy, immunotherapy, surgery or radiotherapy.
  • FIG. 1 shows squamous cell tumor growth with and without oral, intravenous and intratumoral administration of recombinant human lactoferrin.
  • FIG. 2 shows percent tumor growth inhibition in animals receiving lactoferrin, cisplatin and lactoferrin in combination with cisplatin.
  • FIG. 3 shows the percent tumor growth inhibition with lactoferrin in combination with various doses of cisplatin.
  • FIG. 4 shows the NK activity after treatment with lactoferrin.
  • FIG. 5 shows squamous cell tumor growth with and without intratumoral administration of recombinant lactoferrin once or twice a day.
  • hyperproliferative disease refers to any disease or disorder in which the cells proliferate more rapidly than normal tissue growth.
  • a hyperproliferating cell is a cell that is proliferating more rapidly than normal cells.
  • 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, intratumoral, intraocular, or intraarticular administration.
  • intravenous administration includes all techniques to deliver a lactoferrin composition to the systemic circulation via an intravenous injection or infusion.
  • intramoral administration includes all techniques to deliver a lactoferrin composition to the site of a tumor including injection, electroporation, creams, lotions or other forms of administration.
  • oral administration includes oral, buccal, enteral or intragastric administration.
  • topical administration includes application to a dermal, epidermal, subcutaneous or mucosal surface.
  • 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 know 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.
  • lactoferrin or “LF” as used herein refers to native or recombinant lactoferrin.
  • Native lactoferrin can be obtained by 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.
  • subject as used herein, is taken to mean any mammalian subject to which the lactoferrin composition is 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 hyperproliferative disease.
  • terapéuticaally effective amount refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
  • treating refers to administering to a subject a therapeutically effective amount of a 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.
  • improvements in patients with cancer may include tumor stabilization, tumor shrinkage, increased time to progression, increased survival or improvements in the quality of life.
  • Beneficial effect may also be reflected in an improvement of the patient's immune system as measured by the number and activity of circulating immune cells such as CD4+ cells, CD8+ cells, NK cells and CD40+ cells.
  • vicinity refers to in or around the area or site of the tumor and/or hyperproliferative disease.
  • “vicinity of a tumor” may refer to the area in or around the tumor or margins of the tumor. Vicinity includes the area adjacent to the tumor, the area over the tumor, the area under the tumor, the margin area around the tumor, or the area adjacent the tumor margin area.
  • 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 human lactoferrin 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. Pat. Nos. 5,571,896; 5,571,697 and 5,571,691, which are herein incorporated by reference.
  • lactoferrin compositions according to the present invention will be via any common route, orally, parenterally, or topically.
  • exemplary routes include, but are not limited to oral, nasal, buccal, rectal, vaginal, intramuscular, intraperitoneal, intravenous, intraarterial, intratumoral or dermal.
  • Such compositions would normally be administered as pharmaceutically acceptable compositions as described herein.
  • compositions of the present invention may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • Sterile injectable solutions are prepared by incorporating the lactoferrin in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the inventive composition suitable for oral administration is provided in a pharmaceutically acceptable carrier with or without an inert diluent.
  • the carrier should be assimilable or edible and includes liquid, semi-solid, i.e., pastes, or solid carriers. Except insofar as any conventional media, agent, diluent or carrier is detrimental to the recipient or to the therapeutic effectiveness of a lactoferrin preparation contained therein, its use in an orally administrable lactoferrin for use in practicing the methods of the present invention is appropriate.
  • carriers or diluents include fats, oils, water, saline solutions, lipids, liposomes, resins, binders, fillers and the like, or combinations thereof.
  • the composition is combined with the carrier in any convenient and practical manner, i.e., by solution, suspension, emulsification, admixture, encapsulation, microencapsulation, absorption and the like. Such procedures are routine for those skilled in the art.
  • the composition in powder form is combined or mixed thoroughly with a semi-solid or solid carrier.
  • the mixing can be carried out in any convenient manner such as grinding.
  • Stabilizing agents can be also added in the mixing process in order to protect the composition from loss of therapeutic activity through, i.e., denaturation in the stomach.
  • stabilizers for use in an orally administrable composition include buffers, antagonists to the secretion of stomach acids, amino acids such as glycine and lysine, carbohydrates such as dextrose, mannose, galactose, fructose, lactose, sucrose, maltose, sorbitol, mannitol, etc., proteolytic enzyme inhibitors, and the like. More preferably, for an orally administered composition, the stabilizer can also include antagonists to the secretion of stomach acids.
  • composition for oral administration which is combined with a semi-solid 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, i.e., U.S. Pat. No. 5,629,001. Upon reaching the small intestines, the basic pH therein dissolves the coating and permits the composition to be released and absorbed by specialized cells, i.e., epithelial enterocytes and Peyer's patch M cells.
  • specialized cells i.e., epithelial enterocytes and Peyer's patch M cells.
  • 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
  • a specific formulation that may be used in the present invention is a solution of lactoferrin in a hypotonic phosphate based buffer that is free of potassium where the composition of the buffer is as follows: 6 mM sodium phosphate monobasic monohydrate, 9 mM sodium phosphate dibasic heptahydrate, 50 mM sodium chloride, pH 7.0 ⁇ 0.1.
  • the concentration of lactoferrin in a hypotonic buffer may range from 10 microgram/ml to 100 milligram/ml.
  • This formulation may be administered via any route of administration, for example, but not limited to intratumoral administration.
  • a 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.
  • Preferred polymers that are used to manufacture a gel composition of the present invention include, but are not limited to carbopol, carboxymethyl-cellulose, and pluronic polymers.
  • a powdered lactoferrin composition is combined with an aqueous gel containing an polymerization agent such as Carbopol 980 at strengths between 0.5% and 5% wt/volume for application to the skin for treatment of hyperproliferative disease on or beneath the skin.
  • 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 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 Biologics standards.
  • a lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered to a subject suspected of or having a hyperproliferative disease.
  • One of skill in the art can determine the therapeutically effective amount of human lactoferrin to be administered to a subject based upon several considerations, such absorption, metabolism, method of delivery, age, weight, disease severity and response to the therapy.
  • the route of administration will vary, naturally, with the location and nature of the lesion, and include, for example intradermal, transdermal, parenteral, intravenous, intramuscular, intranasal, subcutaneous, percutaneous, intratracheal, intraperitoneal, intratumoral, perfusion, lavage, direct injection, and oral administration.
  • 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.
  • Intratumoral administration of the lactoferrin 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 tumors. Local, regional or systemic administration also may be appropriate.
  • 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, pre-neoplastic 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 100 g of the lactoferrin composition.
  • the lactoferrin composition is orally administered in the range of 1 mg to 100 g per day, more preferably about 20 mg to about 10 g per day with the most preferred dose being 4.5 g per day.
  • Intravenously administered lactoferrin can be in the range of 0.1 ⁇ g to about to 10 g per day, more preferably about 0.1 ⁇ g to about 1 mg with the most preferred dose being 250 mg per day.
  • a lactoferrin composition is intratumorally administered in the range of 0.1 ⁇ g to 10 g per day with the most preferred dose being 100 ⁇ g per day.
  • the amount of lactoferrin may vary from about 1 ⁇ g to about 100 g of lactoferrin.
  • the topical gel, solution, capsule or tablet comprises a lactoferrin concentration of about 0.01% to about 20%. More preferably, the topical gel, solution, capsule or tablet may comprise a lactoferrin concentration of about 1% to about 8.5%.
  • Treatment regimens may vary as well, and often depend on tumor type, tumor location, disease progression, and health and age of the patient. Obviously, certain types of tumor will require more aggressive treatment, while at the same time, certain patients cannot tolerate more taxing protocols. The clinician will be best suited to make such decisions based on the known efficacy and toxicity (if any) of the therapeutic formulations.
  • 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.
  • the lactoferrin 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 lactoferrin 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 further embodiment of the present invention is a method of treating a hyperproliferative disease comprising the step of supplementing a mucosal immune system by increasing the amount of lactoferrin in the gastrointestinal tract.
  • the lactoferrin is administered orally.
  • 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, preferably human lactoferrin.
  • lactoferrin stimulates interleukin-18 and GM-CSF in the gastrointestinal tract, which enhance immune cells.
  • interleukin-18 enhances T lymphocytes or natural killer cells and GM-CSF promotes maturation and migration of immune cells including dendritic and other antigen presenting cells.
  • interleukin-18 IL-18 enhances CD4+, CD8+ and CD3+ cells.
  • IL-18 is a Th1 cytokine that acts in synergy with interleukin-12 and interleukin-2 in the stimulation of lymphocyte IFN-gamma production.
  • Other cytokines may also be enhanced for example, but not limited to IL-1b or, IL-12 or IFN-gamma.
  • lactoferrin 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 lactoferrin in the systemic circulation.
  • the lactoferrin composition is administered intravenously.
  • lactoferrin stimulates interleukin-18 and GM-CSF in the tissue, which enhance immune cells.
  • interleukin-18 enhances T lymphocytes or natural killer cells and GM-CSF promotes maturation and migration of immune cells including dendritic and other antigen presenting cells.
  • interleukin-18 enhances CD4+, CD8+ and CD3+ cells.
  • IL-18 is a Th1 cytokine that acts in synergy with interleukin-12 and interleukin-2 in the stimulation of lymphocyte IFN-gamma production.
  • Other cytokines may also be enhanced for example, but not limited to IL-1b or, IL-12 or IFN-gamma.
  • lactoferrin stimulates 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 lactoferrin stimulates interleukin-18 and GM-CSF in the local tissue, which enhances immune cells.
  • interleukin-18 enhances T lymphocytes or natural killer cells and GM-CSF promotes maturation and migration of immune cells including dendritic and other antigen presenting cells.
  • interleukin-18 enhances CD4+, CD8+ and CD3+ cells.
  • IL-18 is a Th1 cytokine that acts in synergy with interleukin-12 and interleukin-2 in the stimulation of lymphocyte IFN-gamma production.
  • Other cytokines may also be enhanced for example, but not limited to IL-1b or, IL12 or IFN-gamma.
  • lactoferrin 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 lactoferrin in the skin in the vicinity of the tumor.
  • the lactoferrin 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. It is envisioned that lactoferrin stimulates interleukin-18 and GM-CSF in the local tissue (e.g., keratinocytes), which enhances immune cells.
  • interleukin-18 enhances T lymphocytes or natural killer cells and GM-CSF promotes maturation and migration of immune cells including dendritic and other antigen presenting cells.
  • interleukin-18 enhances CD4+, CD8+ and CD3+ cells.
  • IL-18 is a Th1 cytokine that acts in synergy with interleukin-12 and interleukin-2 in the stimulation of lymphocyte IFN-gamma production.
  • Other cytokines may also be enhanced for example, but not limited to EL-1b or, IL-12 or IFN-gamma.
  • lactoferrin stimulates 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. 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 human 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).
  • 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.
  • 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 (VP16), 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 (VP16), 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 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, 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.
  • 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-1, MIP-1beta, MCP-1, 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 effect 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 gp100 or MAGE-3. These antigens are being administered as peptide vaccines and/or as dendritic cell vaccines.
  • Human squamous cell carcinoma (O12) was used. The cells were injected into the right flank of athymic nude mice. rhLF was administered either intratumorally (49 animals, 7 doses ranging from 0.05 ⁇ g to 125 ⁇ g per dose), intravenously (7 animals, 125 ug/dose) or orally (7 animals 20 mg/dose). Control animals were treated with only the vehicle; no rhLF was administered to the control animals. rhLF was administered twice a day for either five days (intravenous group) or eight days (all other groups) starting 11 days after inoculation with tumor cells to allow forrnation of established tumors.
  • Tumor cells from a broad range of tumor types are injected into the right flank of athymic nude mice.
  • Animals are administered either rhLF, native hLF or bovine LF orally.
  • Control animals are treated with only the vehicle, no rhLF is administered to the control animals.
  • rhLF is administered either once or twice a day for either one, five, seven or fourteen days or eight days starting approximately eleven days after inoculation with tumor cells to allow formation of established tumors or at such other time as is generally done with standard or published regimens.
  • the efficacy of treatment is evaluated by measuring the solid tumor size during and at the end of the experiment; the body weights are also determined at the time of tumor measurements.
  • the immune response is measured by measuring the amount of cytokines, T-cells and NK cells in circulation and in the intestine.
  • mice were treated for three days daily with 65 mg/kg/day of rhLF, 300 mg/kg/day of rhLF or 300 mg/kg/day of bLF.
  • mice were only administered the pharmaceutical carrier. Twenty-four hours following administration of the LF or control for 3 days, animals were weighed and blood and serum were collected. Serum was used for cytokine ELISA assays.
  • mice were treated orally with rhLF or placebo once a day for 3 days (see Table 4). TABLE 4 Treatment Regimen Treatment* N Dose (mg/kg) Route Schedule Group 1 Placebo 6 0 — — Group 2 RhLF 7 300 mg/kg/day Oral 3 days
  • mice were sacrificed and spleens were collected. NK cells were separated using a magnetic bead cell sorting assay (MACS anti-NK-DX5) and counted. Cells were then tested in vitro for NK-activity against YAC targets using a lactate dehydrogenase (LDH) release test.
  • LLC lactate dehydrogenase
  • Table 5 shows that oral rhLF treatment resulted in a significant increase of NK activity ex-vivo against YAC-target cells (10%@30:1 versus 2.8% of ctrl group). No significant change in NK activity was observed in placebo treated mice.
  • mice (5 animals per group) were treated for three days daily with 300 mg/kg/day of rhLF. For a control, mice were only administered the pharmaceutical carrier. Twenty-four hours following administration of the LF or placebo for 3 days, animals were and the small intestinal tissue was removed for further analysis. Small intestinal epithelium was homogenized using a lysis buffer consisting of PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulphate containing 10 ⁇ g/ml PhenylMetheylsulfonyl fluoride. Homogenate was centrifuged at 15,000 rpm for 10 minutes and the supernatant stored at ⁇ 80 C. till it was tested for GM-CSF levels using an ELISA kit.
  • a lysis buffer consisting of PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulphate containing 10 ⁇ g/ml PhenylMetheylsulf
  • a murine squamous carcinoma cell line (SCCVII) was injected into the floor of the mouth through the neck skin of immunocompetent C3H mice (Day 0). Five days after tumor cell implantation (Day 5), a skin incision was made in the lower neck and surgical dissection revealed the established tumors. Tumors were measured in three dimensions with calipers.
  • RhLF 4 mg; 200 mg/Kg
  • Cisplatin was administered as a single dose of 5 mg/Kg given intraperitoneally either at the start of rhLF (day 5), in the middle (day 8) or at the end (day 12) of rhLF therapy. Animals were sacrificed on day 12 post-implantation and the residual tumor masses were measured and processed for later additional analyses.
  • RhLF mg/kg Cisplatin Group A Placebo 5 0 (Placebo) 0 Group B RhLF Alone 5 200 mg/kg 0 Group C CP Day 5 5 0 5 mg/kg on day 5* Group D CP Day 8 5 0 5 mg/kg on day 8* Group E CP Day 12 4 0 5 mg/kg on day 12 Group F RhLF/CP-5 5 200 mg/kg 5 mg/kg on day 5* Group G RhLF/CP-8 5 200 mg/kg 5 mg/kg on day 8* Group H RhLF/CP-12 5 200 mg/kg 5 mg/kg on day 12*
  • mice receiving rhLF+cisplatin showed a TGI relative to the relevant group receiving cisplatin alone.
  • animals receiving rhLF+cisplatin showed a 77% TGI relative to the placebo animals (P ⁇ 0.0001), a 66% TGI relative to rhLF alone (P ⁇ 0.01) and a 63% TGI relative to cisplatin alone (P ⁇ 0.01).
  • Cisplatin dosing immediately prior to the start of rhLF (RhLF+CP-5) or during the period of rhLF administration (RhLF+CP-8) provided greater incremental benefit than when cisplatin was administered following completion of rhLF therapy (RhLF+CP-12).
  • RhLF+CP-8 only the straddling regimen (RhLF+CP-8) provided a statistically significant improvement (P ⁇ 0.01) TGI of 77% over cisplatin alone (CP Day 8).
  • TGI Tumor Growth Inhibition
  • SEM Treatment Group Growth Relative to Placebo* Group
  • TGI TGI (%) P-value A (Placebo) 741 (79) — — B (RhLF alone) 496 (155) 33% 0.0989 C (CP Day 5) 240 (137) 68% 0.0066 D (CP Day 8) 693 (146) 6% 0.3898 E (CP Day 12) 433 (175) 42% 0.0634 F (RhLF + CP-5) 14 (5) 98% ⁇ 0.0001 G (RhLF + CP-8) 159 (48) 79% 0.0001 H (RhLF + CP-12) 331 (47) 55% 0.0011 C to E (All CP) 457 (96) 38% 0.0564 F to H (All rhLF/CP) 168 (40) 77% ⁇ 0.0001
  • a murine squamous carcinoma cell line (SCCVII) was injected into the floor of the mouth through the neck skin of immunocompetent C3H mice (Day 0) as described in Example 6. On Day 5 days after initial implantation, tumors were measured for the baseline, then treated with either cisplatin (Day 8, i.p., 5 mg/kg) alone or cisplatin plus three doses of oral rhLF (daily by gavage for 7-8 days on days 5 through 11/12). Animals were sacrificed on Day 11/12 and tumors measured. There was a dose dependent inhibition of tumor growth in the animals receiving both rhLF and cisplatin as compared to the animals receiving cisplatin alone as shown in FIG. 3.
  • SCCVII murine squamous carcinoma cell line
  • a murine squamous carcinoma cell line (SCCVII) was injected into the floor of the mouth through the neck skin of immunocompetent C3H mice (Day 0) as described in Example 7. On Day 5 after initial implantation, tumors were measured for the baseline, then treated with either oral placebo alone (once daily from days 5 to 12; 6 animals), placebo and docetaxel (i.v. bolus of 31.3 mg/kg docetaxel on Day 8; 9 animals), or docetaxel plus oral rhLF (200 mg/kg, administered once daily by gavage from days 5 to 12; 9 animals). Animals were sacrificed on Day 14 and tumors measured.
  • oral placebo alone once daily from days 5 to 12; 6 animals
  • docetaxel i.v. bolus of 31.3 mg/kg docetaxel on Day 8; 9 animals
  • docetaxel plus oral rhLF 200 mg/kg, administered once daily by gavage from days 5 to 12; 9 animals. Animals were sacrificed on Day 14 and tumors measured.
  • a murine squamous carcinoma cell line (SCCVI) was injected into the floor of mouth through the neck skin of immunocompetent C3H mice (Day 0). Five days after tumor cell implantation (Day 5), a skin incision was made in the lower neck and surgical dissection revealed the established tumors. Tumors were measured in three dimensions with calipers.
  • rhLF 200 mg/Kg
  • Radiotherapy was administered as single dose of 2 Gray given at the begirning (day 5) or at during (day 8) rhLF-therapy. Animals were sacrificed on day 14 post-treatment and the residual tumor masses were measured and processed for later additional analyses.
  • mice receiving rhLF alone, radiotherapy alone, or combination therapy showed a significant tumor growth inhibition (TGI) relative to placebo treated mice.
  • TGI tumor growth inhibition
  • TGI Tumor Growth Inhibition
  • lactoferrin stimulated the immune system. Still further, lactoferrin in combination with cisplatin, docetaxel and radiation resulted in inhibition of tumor growth.
  • RhLF Recombinant human lactoferrin was orally administered to human patients with a range of metastatic cancer types that had failed standard chemotherapy in two different studies conducted in multiple centers in four countries (Argentina, Brazil, Chile, U.S.) RhLF was administered at doses of 1.5 to 9 grams daily in two divided doses in cycles of 14 each with a 14 day gap.
  • Tumor size progression was monitored through CT scans and tumor markers where available.
  • CT scans were performed at baseline and after each 8-week period once treatment was initiated, and also compared with a pre-baseline scan conducted prior to enrollment in the study. Tumor markers are measured every 4 weeks.
  • Blood samples were collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples were collected to measure circulating IL-18, IL-1, IL-2, and IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Table 12 shows the tumor response of five individual patients with different tumor types. In all cases, the percent growth of the tumor size prior to treatment of rhLF (the relevant duration of time is shown in parentheses) and the growth of the tumor in the ensuing two time periods, as measured by CT, showed a diminution in their rate of tumor growth or an actual shrinkage.
  • Recombinant human lactoferrin is orally administered to human patients to inhibit tumor growth either alone or in combination with standard anti-cancer regimens.
  • rhLF is administered using the optimum regimen and doses identified in Example 10 and the standard anti-cancer regimen(s) for the selected tumor type is used as part of the combination therapy.
  • the route of administration and regimen of the additional anti-cancer therapy is as approved by the FDA for that indication or as described in a peer reviewed publication.
  • Tumor size progression is monitored through CT scans and tumor markers where available.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks once treatment is initiated.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, and IL-12 and IFN- ⁇ .
  • mice were treated orally with rhLF or placebo once a day for 3 days. One day later (day 4), mice were sacrificed and spleens collected. NK cells were separated using a magnetic bead cell sorting assay (MACS anti-NK-DX5) and counted. Cells were then tested in vitro for NK-activity against YAC targets using a lactate dehydrogenase (LDH) release test.
  • LLC lactate dehydrogenase
  • Recombinant lactoferrin, bovine lactoferrin and native lactoferrin are intravenously administered to animals, preferably rats, and the production of IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN-gamma in the plasma, serum and blood packed cells are measured.
  • rats are treated for fourteen consecutive days with 0.05 ⁇ g to 1000 ⁇ g per dose.
  • rats are only administered the pharmaceutical carrier.
  • animals are weighed and blood and serum are collected.
  • the levels of CD4+, CD8+ and NK cells are counted from the blood that was collected.
  • Plasma, serum and an extract of the blood cells are used for cytokine ELISA assays.
  • tissue are removed for further analysis. Tissues are homogenized using a lysis buffer consisting of PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulphate containing 10 ⁇ g/ml PhenylMetheylsulfonyl fluoride. Homogenate is centrifuged at 15,000 rpm for 10 minutes and the supernatant stored at ⁇ 80 C. till it is tested for the cytokines IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN-gamma.
  • a lysis buffer consisting of PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulphate containing 10 ⁇ g/ml PhenylMetheylsulfonyl fluoride. Homogenate is centrifuged at 15,000 rpm for 10 minutes and the supernatant stored at ⁇ 80
  • Tumor cells to be tested are injected into the right flank of athymic nude mice. Animals are administered rhLF intravenously alone and in combination with other anti-cancer regimens as described in Example 13. Control animals are treated with only the vehicle; no rhLF is administered to the control animals. rhLF is administered using regimens identified as being optimal in the trials described in Example 13. Anti-cancer therapy is administered using standard or published regimens. Therapy starts approximately 11 days after inoculation with tumor cells to allow formation of established tumors or at such other time as is generally done with standard or published regimens.
  • Recombinant lactoferrin is intravenously administered to patients to inhibit tumor growth.
  • rhLF at a dose of 500 mg per day for eight days to patients suffering from unresectable or metastatic cancer.
  • rhLF is administered for one to eight days to patients suffering from metastatic cancer in daily doses of 0.1, 1, 10, 100, and 1000 mg.
  • the dose is administered intravenously.
  • Tumor size progression is monitored through CT scans and tumor markers where available.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Recombinant lactoferrin is intravenously administered to patients to inhibit tumor growth either alone or in combination with standard anti-cancer regimens.
  • rhLF is administered using the optimum regimen and doses identified in Example 15 and the standard anti-cancer regimen(s) for the selected tumor type is used as part of the combination therapy.
  • the route of administration and regimen of the additional anti-cancer therapy is as approved by the FDA for that indication or as described in a peer reviewed publication.
  • Tumor size progression is monitored through CT scans and tumor markers where available.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks once treatment is initiated.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • O12 human oropharyngeal squamous cell carcinoma tumor cells were injected to the right flank of athymic nude mice.
  • Recombinant human lactoferrin and vehicle controls were dosed via intratumoral injection.
  • Each animal was administered different concentrations of rhLF in 50 ⁇ L doses consisting of four separate injections of approximately 12.5 ⁇ L of the dose, at different directions and angles (approximately S/N/E/W) to ensure that the dose was distributed evenly throughout the tumor (fanning).
  • Table 13 shows the regimen followed for each experimental group and the dose of rhLF per injection for each animal per group.
  • rhLF was administered directly into the tumor.
  • Each animal was tracked daily for tumor growth by external caliper measurements of the protruding tumor.
  • mice Normal C3H/HeJ mice were implanted with one of two mouse tumors following the methodology described in Example 17. Tumors used were SCCVII and RIF mouse tumor cell lines. Following establishment of the tumors in the mice, tumors were injected intratumorally daily for 4 days with 250 or 500 ⁇ g rhLF per dose or with vehicle control. Twenty four hours following the last intratumoral injection, animals were sacrificed and the blood examined for lymphocyte populations. The number of circulating lymphocytes were increased by 34% to 56% relative to the placebo treated control animals (Table 14).
  • Tumor cells to be tested are injected into the right flank of athymic nude mice. Animals are administered rhLF intratumorally alone and in combination with other anti-cancer regimens as described in Example 1 or Example 17. Control animals are treated with only the vehicle; no rhLF is administered to the control animals.
  • Anti-cancer therapy is administered using standard or published regimens. Therapy starts approximately 11 days after inoculation with tumor cells to allow formation of established tumors or at such other time as is generally done with standard or published regimens.
  • Recombinant lactoferrin is intratumorally administered to patients to inhibit tumor growth.
  • rhLF at a dose of 1000 ⁇ g per day for eight days to patients suffering from unresectable or metastatic cancer.
  • rhLF is administered for one to eight days to patients suffering from metastatic cancer in daily doses of 10, 50, 100, 500 and 1000 ⁇ g.
  • the dose is administered intratumorally.
  • Tumor size progression is monitored through CT scans and tumor markers where available.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Recombinant lactoferrin is intratumorally administered to patients to inhibit tumor growth either alone or in combination with standard anti-cancer regimens.
  • rhLF is administered using the optimum regimen and doses identified in Example 20 and the standard anti-cancer regimen(s) for the selected tumor type is used as part of the combination therapy.
  • the route of administration and regimen of the additional anti-cancer therapy is as approved by the FDA for that indication or as described in a peer reviewed publication.
  • Tumor size progression is monitored through CT scans and tumor markers where available.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks once treatment is initiated.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Recombinant lactoferrin in a gel formulation is administered to patients to inhibit tumor growth.
  • rhLF gel at strengths of 1%, 2.5% or 8.5% is applied twice a day to a skin or subcutaneous cancerous lesion in a patient with metastatic disease. Application of rhLF gel continues till tumor progression.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Recombinant lactoferrin in a gel formulation is administered to patients to inhibit tumor growth either alone or in combination with standard anti-cancer regimens.
  • rhLF is administered using the optimum regimen and doses identified in Examples 22 and the standard anti-cancer regimen(s) for the selected tumor type is used as part of the combination therapy.
  • the route of administration and regimen of the additional anti-cancer therapy is as approved by the FDA for that indication or as described in a peer reviewed publication.
  • CT scans are performed at baseline and after each 8-week period once treatment is initiated. Tumor markers are measured every 4 weeks once treatment is initiated.
  • Blood samples are collected to measure subclasses of circulating lymphocytes and NK cell activity. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • Gahr M et al., J Leukocyte Biol. 1991;49: 427-33.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cardiology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Vascular Medicine (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US10/435,319 2002-05-10 2003-05-09 Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases Abandoned US20040082504A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/435,319 US20040082504A1 (en) 2002-05-10 2003-05-09 Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US12/561,429 US20100137208A1 (en) 2002-05-10 2009-09-17 Intratumorally administered lactoferrin in the treatment of malignantneoplasms and other hyperproliferative diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US37947402P 2002-05-10 2002-05-10
US37944102P 2002-05-10 2002-05-10
US37944202P 2002-05-10 2002-05-10
US10/435,319 US20040082504A1 (en) 2002-05-10 2003-05-09 Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/561,429 Continuation US20100137208A1 (en) 2002-05-10 2009-09-17 Intratumorally administered lactoferrin in the treatment of malignantneoplasms and other hyperproliferative diseases

Publications (1)

Publication Number Publication Date
US20040082504A1 true US20040082504A1 (en) 2004-04-29

Family

ID=29424524

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/435,319 Abandoned US20040082504A1 (en) 2002-05-10 2003-05-09 Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US10/434,769 Expired - Fee Related US7901879B2 (en) 2002-05-10 2003-05-09 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US12/561,429 Abandoned US20100137208A1 (en) 2002-05-10 2009-09-17 Intratumorally administered lactoferrin in the treatment of malignantneoplasms and other hyperproliferative diseases
US12/964,327 Expired - Fee Related US8242079B2 (en) 2002-05-10 2010-12-09 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US13/546,208 Abandoned US20120276126A1 (en) 2002-05-10 2012-07-11 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases

Family Applications After (4)

Application Number Title Priority Date Filing Date
US10/434,769 Expired - Fee Related US7901879B2 (en) 2002-05-10 2003-05-09 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US12/561,429 Abandoned US20100137208A1 (en) 2002-05-10 2009-09-17 Intratumorally administered lactoferrin in the treatment of malignantneoplasms and other hyperproliferative diseases
US12/964,327 Expired - Fee Related US8242079B2 (en) 2002-05-10 2010-12-09 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US13/546,208 Abandoned US20120276126A1 (en) 2002-05-10 2012-07-11 Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases

Country Status (12)

Country Link
US (5) US20040082504A1 (zh)
EP (2) EP1507554B1 (zh)
JP (2) JP4685443B2 (zh)
CN (2) CN100467060C (zh)
AT (1) ATE519499T1 (zh)
AU (2) AU2003239393A1 (zh)
CA (1) CA2484656A1 (zh)
CY (1) CY1111999T1 (zh)
DK (1) DK1507554T3 (zh)
HK (2) HK1080722A1 (zh)
PT (1) PT1507554E (zh)
WO (2) WO2003094952A1 (zh)

Cited By (9)

* 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
US20040009896A1 (en) * 2002-05-24 2004-01-15 Peter Glynn Oral lactoferrin in the treatment of respiratory disorders
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US20040152624A1 (en) * 2002-12-06 2004-08-05 Atul Varadhachary Oral lactoferrin in the treatment of sepsis
US20040176276A1 (en) * 2002-12-10 2004-09-09 Atul Varadhachary Lactoferrin as an agent in the prevention of organ transplant rejection and graft-versus-host-disease
US20050019342A1 (en) * 2003-06-06 2005-01-27 Agennix Incorporated Lactoferrin as an adjuvant in cancer vaccines
US20050075277A1 (en) * 2003-07-10 2005-04-07 Agennix Incorporated Use of lactoferrin in prophylaxis against infection and/or inflammation in immunosuppressed subjects
US20060094082A1 (en) * 2004-10-26 2006-05-04 Agennix Incorporated Composition of lactoferrin related peptides and uses thereof
US7244706B2 (en) 1999-02-05 2007-07-17 Agennix, Inc. Antimicrobial/endotoxin neutralizing polypeptide

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7323443B2 (en) * 2002-12-12 2008-01-29 Agennix Inc. Lactoferrin in the reduction of pain
GB0413954D0 (en) * 2004-06-22 2004-07-28 Altunkaya Ali Compositions for topical treatment
EP1835930A4 (en) * 2004-11-19 2012-11-21 Fonterra Corporate Res And Dev Ltd PROCESS FOR HEMATOLOGIC OR IMMUNE IMPROVEMENT, PREVENTING TUMOR FORMATION OR TUMOR GROWTH AND TREATMENT OR PREVENTION OF CANCER
WO2007022537A2 (en) * 2005-08-19 2007-02-22 Agennix Incorporated Use of lactoferrin as a chemokine and a chemotactic modulator
BRPI0713642A2 (pt) * 2006-06-14 2012-10-23 Nutricia Nv composição anti-inflamatória, e, uso de uma composição
WO2010033424A2 (en) * 2008-09-19 2010-03-25 Nestec S.A. Nutritional support of the immune system during anti-cancer treatment
EP2391225B1 (en) * 2009-01-28 2020-05-06 Jean-Paul Perraudin Method for production of lactoferrin
US10314594B2 (en) 2012-12-14 2019-06-11 Corquest Medical, Inc. Assembly and method for left atrial appendage occlusion
US10813630B2 (en) 2011-08-09 2020-10-27 Corquest Medical, Inc. Closure system for atrial wall
US10307167B2 (en) 2012-12-14 2019-06-04 Corquest Medical, Inc. Assembly and method for left atrial appendage occlusion
US20140142689A1 (en) 2012-11-21 2014-05-22 Didier De Canniere Device and method of treating heart valve malfunction
JP2015067560A (ja) * 2013-09-27 2015-04-13 国立大学法人広島大学 ラクトフェリンを含有する癌転移抑制剤
US9566443B2 (en) 2013-11-26 2017-02-14 Corquest Medical, Inc. System for treating heart valve malfunction including mitral regurgitation
US9895410B2 (en) 2013-12-12 2018-02-20 Cornell University Methods for preventing and treating oral cancers
US10842626B2 (en) 2014-12-09 2020-11-24 Didier De Canniere Intracardiac device to correct mitral regurgitation
CN106174462A (zh) * 2016-04-17 2016-12-07 马鞍山市志诚科技有限公司 一种防治血管瘤的营养配方纳米颗粒及制备加工方法
JPWO2018079701A1 (ja) * 2016-10-28 2019-09-19 株式会社Nrlファーマ ラクトフェリン活性を有するタンパク質を含む、抗ガン治療補助剤
CN111544579A (zh) * 2020-03-13 2020-08-18 中国农业科学院北京畜牧兽医研究所 一种抗癌症的药物组合物

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4977137A (en) * 1987-06-03 1990-12-11 Baylor College Of Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US5198419A (en) * 1989-12-08 1993-03-30 Immuno Japan Inc. Formulated medicines for enhancing the efficacy of beta-lactam antibiotics in prophylaxis and treatment against infectious disease due to pathogenic bacteria
US5571697A (en) * 1989-05-05 1996-11-05 Baylor College Of Medicine Texas Medical Center Expression of processed recombinant lactoferrin and lactoferrin polypeptide fragments from a fusion product in Aspergillus
US5571691A (en) * 1989-05-05 1996-11-05 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using CDNA sequences in various organisms
US5571896A (en) * 1992-04-24 1996-11-05 Baylor College Of Medicine Production of recombinant human lactoferrin
US5766939A (en) * 1989-05-05 1998-06-16 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using CDNA sequences in various organisms
US5849881A (en) * 1989-05-05 1998-12-15 Baylor College Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US6100054A (en) * 1989-05-05 2000-08-08 Baylor College Of Medicine Production for recombinant lactoferrin and lactoferrin polypeptides using DNA sequences in various organisms
US6111081A (en) * 1996-05-31 2000-08-29 Baylor College Of Medicine Lactoferrin variants and uses thereof
US6228614B1 (en) * 1989-05-05 2001-05-08 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US6333311B1 (en) * 1997-02-03 2001-12-25 Pharming Useful properties of human lactoferrin and variants thereof
US6399570B1 (en) * 1999-02-05 2002-06-04 Agennix, Inc. Antimicrobial/endotoxin neutralizing polypeptide
US20030022821A1 (en) * 1998-08-28 2003-01-30 Alpharma As Bioactive peptides
US20030096736A1 (en) * 2001-05-09 2003-05-22 Kruzel Marian L. Lactoferrin for age related disorders in humans
US20030190303A1 (en) * 1997-04-10 2003-10-09 Ian Kimber Lactoferrin as regulator of allergen-induced tumor necrosis factor-alpha production and therapeutic applications
US20040009896A1 (en) * 2002-05-24 2004-01-15 Peter Glynn Oral lactoferrin in the treatment of respiratory disorders
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
US20050075277A1 (en) * 2003-07-10 2005-04-07 Agennix Incorporated Use of lactoferrin in prophylaxis against infection and/or inflammation in immunosuppressed subjects

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6351337A (ja) * 1986-08-22 1988-03-04 Snow Brand Milk Prod Co Ltd 抗腫瘍剤
ATE158180T1 (de) 1991-06-21 1997-10-15 Univ Cincinnati Oral verabreichbare therapeutische proteine und herstellungsverfahren
JP3184923B2 (ja) * 1992-01-08 2001-07-09 ビオ セレ ラボラトワール エス ア 抗リウマチ剤
CA2086874E (en) * 1992-08-03 2000-01-04 Renzo Mauro Canetta Methods for administration of taxol
US5679807A (en) * 1995-01-30 1997-10-21 Hauser, Inc. Preparation of taxol and docetaxel through primary amines
JPH08217693A (ja) * 1995-02-17 1996-08-27 Yoshihisa Naito 新規医薬組成物
JP3888707B2 (ja) * 1996-01-22 2007-03-07 森永乳業株式会社 血管新生病治療剤
JP3496387B2 (ja) 1996-01-23 2004-02-09 花王株式会社 毛髪化粧料
WO1998006425A1 (en) 1996-08-12 1998-02-19 A+ Science Invest Ab Treatment and prevention of infections, inflammations and/or tumours with lactoferrin and/or lactoferricin
NZ509101A (en) * 1998-06-26 2003-07-25 N Pharmaceutical preparations for use in combatting or preventing surface infections caused by microorganisms
JP2000229881A (ja) 1999-02-10 2000-08-22 Morinaga Milk Ind Co Ltd 癌疾患の予後改善剤
RU2165769C1 (ru) 2000-07-13 2001-04-27 Якубовская Раиса Ивановна Антибактериальный, антиоксидантный, иммуномодулирующий и антиканцерогенный препарат и способ его применения
AU2004289170B2 (en) * 2003-06-06 2009-12-17 Agennix Incorporated Lactoferrin as an adjuvant in cancer vaccines
EP1835930A4 (en) 2004-11-19 2012-11-21 Fonterra Corporate Res And Dev Ltd PROCESS FOR HEMATOLOGIC OR IMMUNE IMPROVEMENT, PREVENTING TUMOR FORMATION OR TUMOR GROWTH AND TREATMENT OR PREVENTION OF CANCER
JP2007233064A (ja) 2006-03-01 2007-09-13 Bitsign:Kk 表示システム及び表示器

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4977137B1 (en) * 1987-06-03 1994-06-28 Baylor College Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US4977137A (en) * 1987-06-03 1990-12-11 Baylor College Of Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US6228614B1 (en) * 1989-05-05 2001-05-08 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US6080559A (en) * 1989-05-05 2000-06-27 Agennix, Inc. Expression of processed recombinant lactoferrin and lactoferrin polypeptide fragments from a fusion product in Aspergillus
US6635447B1 (en) * 1989-05-05 2003-10-21 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US20050064546A1 (en) * 1989-05-05 2005-03-24 Conneely Orla M. Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US5766939A (en) * 1989-05-05 1998-06-16 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using CDNA sequences in various organisms
US5849881A (en) * 1989-05-05 1998-12-15 Baylor College Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US5955316A (en) * 1989-05-05 1999-09-21 Agennix, Inc. Expression of processed recombinant lactoferrin and lactoferrin polypeptide fragments from a fusion product in aspergillus
US5571697A (en) * 1989-05-05 1996-11-05 Baylor College Of Medicine Texas Medical Center Expression of processed recombinant lactoferrin and lactoferrin polypeptide fragments from a fusion product in Aspergillus
US6100054A (en) * 1989-05-05 2000-08-08 Baylor College Of Medicine Production for recombinant lactoferrin and lactoferrin polypeptides using DNA sequences in various organisms
US5571691A (en) * 1989-05-05 1996-11-05 Baylor College Of Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using CDNA sequences in various organisms
US5198419A (en) * 1989-12-08 1993-03-30 Immuno Japan Inc. Formulated medicines for enhancing the efficacy of beta-lactam antibiotics in prophylaxis and treatment against infectious disease due to pathogenic bacteria
US5571896A (en) * 1992-04-24 1996-11-05 Baylor College Of Medicine Production of recombinant human lactoferrin
US6111081A (en) * 1996-05-31 2000-08-29 Baylor College Of Medicine Lactoferrin variants and uses thereof
US6333311B1 (en) * 1997-02-03 2001-12-25 Pharming Useful properties of human lactoferrin and variants thereof
US20030190303A1 (en) * 1997-04-10 2003-10-09 Ian Kimber Lactoferrin as regulator of allergen-induced tumor necrosis factor-alpha production and therapeutic applications
US20030022821A1 (en) * 1998-08-28 2003-01-30 Alpharma As Bioactive peptides
US20030105006A1 (en) * 1999-02-05 2003-06-05 Mann David M. Antimicrobial/endotoxin neutralizing polypeptide
US6399570B1 (en) * 1999-02-05 2002-06-04 Agennix, Inc. Antimicrobial/endotoxin neutralizing polypeptide
US20030096736A1 (en) * 2001-05-09 2003-05-22 Kruzel Marian L. Lactoferrin for age related disorders in humans
US20040009895A1 (en) * 2002-05-10 2004-01-15 Atul Varadhachary Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040009896A1 (en) * 2002-05-24 2004-01-15 Peter Glynn Oral lactoferrin in the treatment of respiratory disorders
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US20050075277A1 (en) * 2003-07-10 2005-04-07 Agennix Incorporated Use of lactoferrin in prophylaxis against infection and/or inflammation in immunosuppressed subjects

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7244706B2 (en) 1999-02-05 2007-07-17 Agennix, Inc. Antimicrobial/endotoxin neutralizing polypeptide
US7901879B2 (en) * 2002-05-10 2011-03-08 Agennix Incorporated Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040009895A1 (en) * 2002-05-10 2004-01-15 Atul Varadhachary Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US8242079B2 (en) 2002-05-10 2012-08-14 Agennix Incorporated Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040009896A1 (en) * 2002-05-24 2004-01-15 Peter Glynn Oral lactoferrin in the treatment of respiratory disorders
US7238661B2 (en) 2002-05-24 2007-07-03 Agennix, Inc. Oral lactoferrin in the treatment of respiratory disorders
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US7524814B2 (en) 2002-09-16 2009-04-28 Agennix, Inc. Lactoferrin compositions and methods of wound treatment
US8030272B2 (en) 2002-09-16 2011-10-04 Agennix Incorporated Lactoferrin compositions and methods of wound treatment
US8247373B2 (en) 2002-09-16 2012-08-21 Agennix Incorporated Lactoferrin compositions and methods of wound treatment
US20090253627A1 (en) * 2002-09-16 2009-10-08 Agennix Incorporated Lactoferrin compositions and methods of wound treatment
US20040152624A1 (en) * 2002-12-06 2004-08-05 Atul Varadhachary Oral lactoferrin in the treatment of sepsis
US20100210510A1 (en) * 2002-12-06 2010-08-19 Agennix Inc Oral lactoferrin in the treatment of sepsis
US20040176276A1 (en) * 2002-12-10 2004-09-09 Atul Varadhachary Lactoferrin as an agent in the prevention of organ transplant rejection and graft-versus-host-disease
US7592306B2 (en) 2002-12-10 2009-09-22 Agennix, Inc. Lactoferrin as an agent in the prevention of organ transplant rejection and graft-versus-host-disease
US8058234B2 (en) 2002-12-10 2011-11-15 Agennix Incorporated Lactoferrin as an agent in the prevention of organ transplant rejection and graft-versus-host-disease
US8105615B2 (en) 2003-06-06 2012-01-31 Agennix Incorporated Lactoferrin as an adjuvant in cancer vaccines
US20050019342A1 (en) * 2003-06-06 2005-01-27 Agennix Incorporated Lactoferrin as an adjuvant in cancer vaccines
US20110110983A1 (en) * 2003-06-06 2011-05-12 Atul Varadhachary Lactoferrin as an adjuvant in cancer vaccines
US20050075277A1 (en) * 2003-07-10 2005-04-07 Agennix Incorporated Use of lactoferrin in prophylaxis against infection and/or inflammation in immunosuppressed subjects
US7420033B2 (en) 2004-10-26 2008-09-02 Agennix, Inc. Composition of lactoferrin related peptides and uses thereof
US20070142292A1 (en) * 2004-10-26 2007-06-21 Agennix Incorporated Composition of lactoferrin related peptides and uses thereof
US7183381B2 (en) 2004-10-26 2007-02-27 Agennix, Inc. Composition of lactoferrin related peptides and uses thereof
US20060094082A1 (en) * 2004-10-26 2006-05-04 Agennix Incorporated Composition of lactoferrin related peptides and uses thereof

Also Published As

Publication number Publication date
CN1668325A (zh) 2005-09-14
US20110076295A1 (en) 2011-03-31
AU2003239393A1 (en) 2003-11-11
EP1507554B1 (en) 2011-08-10
CN1668326A (zh) 2005-09-14
HK1080722A1 (en) 2006-05-04
WO2003094952A1 (en) 2003-11-20
US7901879B2 (en) 2011-03-08
US20100137208A1 (en) 2010-06-03
JP4685443B2 (ja) 2011-05-18
HK1083203A1 (en) 2006-06-30
JP2005533029A (ja) 2005-11-04
AU2003273182B2 (en) 2009-04-23
DK1507554T3 (da) 2011-11-21
CN100467060C (zh) 2009-03-11
CA2484656A1 (en) 2003-12-04
WO2003099323A1 (en) 2003-12-04
JP2011079858A (ja) 2011-04-21
CY1111999T1 (el) 2015-11-04
EP1507554A1 (en) 2005-02-23
AU2003273182A1 (en) 2003-12-12
JP5156084B2 (ja) 2013-03-06
ATE519499T1 (de) 2011-08-15
US20120276126A1 (en) 2012-11-01
US20040009895A1 (en) 2004-01-15
CN100467059C (zh) 2009-03-11
EP2286827A1 (en) 2011-02-23
PT1507554E (pt) 2011-11-21
EP1507554A4 (en) 2007-05-09
US8242079B2 (en) 2012-08-14
AU2003273182B8 (en) 2009-05-07

Similar Documents

Publication Publication Date Title
US8242079B2 (en) Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
EP2344148B2 (en) Nutritional support to prevent or moderate bone marrow paralysis during anti-cancer treatment
JP6185500B2 (ja) 癌性腫瘍からの骨髄毒性を阻止又は軽減するための栄養支援
US20110110983A1 (en) Lactoferrin as an adjuvant in cancer vaccines
JP4379924B2 (ja) レクチン組成物およびその使用
US6479068B1 (en) Therapeutic nutrient regimen for alleviating mucositis, stomatitis and cachexia in oncology patients
ES2368214T3 (es) Lactoferrina en el tratamiento de neoplasmas malignos y de otras enfermedades hiperproliferativas.
Akmansu et al. The effect of using oral glutamine on the side effect of mucositis in patients with head and neck cancer who are receiving chemoradiotherapy: retrospective evaluation with clinical and immunological parameters
Cribb Whey proteins and immunity

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGENNIX INCORPORATED, TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VARADHACHARY, ATUL;BARSKY, RICK;PETRAK, KAREL;REEL/FRAME:014589/0380

Effective date: 20030813

AS Assignment

Owner name: GPC BIOTECH AG, NEW JERSEY

Free format text: SECURITY AGREEMENT;ASSIGNOR:AGENNIX INCORPORATED;REEL/FRAME:022399/0104

Effective date: 20090217

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: AGENNIX INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:O'MALLEY, BERT W., JR;REEL/FRAME:023307/0497

Effective date: 20071113

Owner name: AGENNIX INC.,TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:O'MALLEY, BERT W., JR;REEL/FRAME:023307/0497

Effective date: 20071113