US20110110983A1 - Lactoferrin as an adjuvant in cancer vaccines - Google Patents

Lactoferrin as an adjuvant in cancer vaccines Download PDF

Info

Publication number
US20110110983A1
US20110110983A1 US12/555,296 US55529609A US2011110983A1 US 20110110983 A1 US20110110983 A1 US 20110110983A1 US 55529609 A US55529609 A US 55529609A US 2011110983 A1 US2011110983 A1 US 2011110983A1
Authority
US
United States
Prior art keywords
lactoferrin
cancer
composition
terminal
subject
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
US12/555,296
Inventor
Atul Varadhachary
Federica Pericle
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 US12/555,296 priority Critical patent/US20110110983A1/en
Publication of US20110110983A1 publication Critical patent/US20110110983A1/en
Assigned to AGENNIX INCORPORATED reassignment AGENNIX INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PERICLE, FEDERICA, VARADHACHARY, ATUL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • 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
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides

Definitions

  • the present invention relates to methods of treating cancer by administering a composition of lactoferrin (LF) in combination with cancer vaccines.
  • LF lactoferrin
  • 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).
  • Immunotherapy includes both innate and specific immune responses that have the potential to treat different tumor types.
  • the activation of tumor antigen-specific T lymphocytes or non-specific macrophages and natural killer (NK) cells using immunotherapeutic approaches may lead to the destruction of tumor cells (Curiel et al., 2002).
  • Cancer vaccines involve the induction of a specific immune response.
  • the administration of a tumor antigen alone is often not sufficient to stimulate an appropriate immune response.
  • Incorporating an immunological adjuvant into a vaccine regimen often improves anti-tumor activity (Dredge et al., 2002).
  • 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 stimulate lymphokine-activated killer
  • Recombinant human lactoferrin has previously been described as being purified after expression in a variety of prokaryotic and eukaryotic organisms including aspergillus (U.S. Pat. No. 6,080,559), cattle (U.S. Pat. No. 5,919,913), rice, corn, Sacharomcyes (U.S. Pat. No. 6,228,614) and Pichia pastoris (U.S. Pat. No. 6,455,687, 6,277,817, 6,066,469). Also described are expression systems for the expression of full-length human lactoferrins (e.g., U.S. Pat. No. 6,100,054).
  • 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 cancer vaccine adjuvant for the prevention (prophylaxis) or treatment (therapeutic) of tumors.
  • the present invention is directed to a method for preventing or treating cancer.
  • the method of treatment involves administration of lactoferrin and a cancer immunotherapy, e.g., cancer vaccine.
  • lactoferrin can be used as an adjuvant for cancer immunotherapy.
  • the lactoferrin composition which is dispersed in a pharmaceutically acceptable carrier, comprises lactoferrin or an N-terminal lactoferrin variant in which at least the N-terminal glycine residue is truncated or substituted.
  • the lactoferrin is mammalian lactoferrin, more particularly, the lactoferrin is human or bovine. Yet further, the lactoferrin is recombinant lactoferrin.
  • N-terminal lactoferrin variants include variants that at least lack the N-terminal glycine residue or contain a substitution at the N-terminal glycine residue. The substitution can comprise substituting a natural or artificial amino acid residue for the N-terminal glycine residue.
  • the substitution can comprise substituting a positive amino acid residue or a negative amino acid residue for the N-terminal glycine residue or substituting a neutral amino acid residue other than glycine for the N-terminal glycine residue.
  • Other N-terminal lactoferrin variants include lactoferrin lacking one or more N-terminal residues or having one or more substitutions in the N-terminal.
  • the 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 amount of the lactoferrin that is administered is about 1 mg to about 100 g per day, more preferably, the amount is about 10 mg to about 100 g per day. More particularly, the composition is a solution, capsule or a tablet having a lactoferrin concentration of about 0.01% to about 100%.
  • Another embodiment of the present invention comprises a method of treating cancer comprising the step of administering to a subject a cancer immunotherapy and an adjuvant, wherein the adjuvant is a lactoferrin composition that is administered in an amount sufficient to provide an improvement in the cancer in the subject. Still further, the method can further comprise additionally administering chemotherapy, immunotherapy, surgery, biotherapy, radiotherapy or a combination thereof to the subject.
  • the lactoferrin composition can be administered orally, parenterally or topically.
  • Parenterally includes, but is not limited subcutaneously, intramuscularly, intraperitoneally, intravenously, intraarterially, intramyocardially, transendocardially, transepicardially, or intrathecally.
  • an antacid in combination with the lactoferrin composition can be administered.
  • the lactoferrin composition can be formulated in a delayed release formulation. Still further, the lactoferrin composition can be formulated wherein release occurs in the small intestine or in the large intestine.
  • the cancer 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. More particularly, the neoplasm is a hematopoietic neoplasm.
  • Exemplary hematopoietic neoplasms include, but are not limited to myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, and chronic lymphocytic leukemia.
  • the immunotherapy comprises antigen presenting cells. More specifically, the lactoferrin composition is administered ex vivo to the antigen presenting cells prior to administering the cells to the subject.
  • the cells are allogeneic or syngeneic.
  • the immunotherapy comprises administration of a tumor antigen to the subject, or administration of a nucleic acid sequence expressing a cancer antigen to the subject.
  • the nucleic acid sequence is contained in a vector.
  • the immunotherapy comprises administration of a vector containing a nucleic acid sequence expressing an immunomodulatory cytokine to the subject.
  • the immunotherapy comprises administration of a protein or nucleic acid that promotes the recognition of a cancer antigen in the subject.
  • the lactoferrin composition is administered simultaneously and/or sequentially with the immunotherapy.
  • Another method of the present invention comprises a method of enhancing the immune system in a subject suffering from cancer or susceptible to cancer comprising the step of administering to the subject a cancer immunotherapy and an adjuvant, wherein the adjuvant is a lactoferrin composition. More specifically, the lactoferrin is administered orally.
  • lactoferrin stimulates the production of interleukin-18, GM-CSF, or MIP-3alpha.
  • interleukin-18, GM-CSF or MIP-3alpha stimulate the production, maturation, migration or activity of immune cells (e.g., T lymphocytes, natural killer cells, dendritic cells, antigen presenting cells or progenitor cell). More specifically, T lymphocytes are selected from the group consisting of CD4+, CD8+ and CD3+ cells.
  • FIG. 1 shows carcinoma cell tumor growth with and without oral administration of lactoferrin.
  • FIG. 2 shows the effect of rhLF on mucosal immunity in mice.
  • FIG. 3 shows and ECD-TM vaccination and oral LF schedule.
  • FIG. 4A and FIG. 4B show the effect of oral LF in combination with ECD-TM vaccination n carcinogenesis in BALB-neuT mice.
  • adjuvant refers to substance added to drug product formulation that affects the action of the active ingredient by enhancing or potentiating its activity.
  • An adjuvant also includes a treatment and/or therapy that is added or combined with a traditional treatment and/or therapy to enhance or potentiate or extend the effect of the traditional treatment and/or therapy.
  • allogeneic refers to cell types or tissues that are antigenically distinct. Thus, cells or tissue transferred from the same species can be antigenically distinct.
  • antigen as used herein is defined as a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both.
  • An antigen can be derived from organisms, subunits of proteins/antigens, killed or inactivated whole cells or lysates. Therefore, a skilled artisan realizes that any macromolecule, including virtually all proteins or peptides, can serve as antigens. Furthermore, antigens can be derived from recombinant or genomic DNA.
  • any DNA which contains nucleotide sequences or partial nucleotide sequences of a pathogenic genome or a gene or a fragment of a gene for a protein that elicits an immune response results in synthesis of an antigen.
  • antigen presenting cell is any cell that enhances the immune response (i.e., from the T-cell or —B-cell arms of the immune system) against an antigen or antigenic composition.
  • dendritic cell or “DC” as used herein is defined as an example of an antigen presenting cell derived from bone marrow. Dendritic cells have a branched or dendritic morphology and are the most potent stimulations of T-cell response.
  • ex vivo refers to “outside” the body.
  • ex vivo and in vitro can be used interchangeably.
  • 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.
  • 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 composition refers to a composition having lactoferrin, a portion or part of lactoferrin, an N-terminal lactoferrin variant, or a combination thereof.
  • 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.
  • human lactoferrin or “hLF” as used herein refers to native or recombinant human lactoferrin.
  • Native human lactoferrin can be obtained by purification from human milk or colostrum or from other natural sources.
  • Recombinant human lactoferrin rhLF
  • rhLF 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.
  • bovine lactoferrin or “bLF’ as used herein refers to native or recombinant bovine lactoferrin.
  • Native bovine lactoferrin can be obtained by purification from bovine milk.
  • Recombinant bovine lactoferrin (rbLF) 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.
  • 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 (e.g., IL-18, GM-CSF or IFN- ⁇ ) by inhibiting various cytokines, (e.g., IL-2, IL-4, IL-5, IL-10, or TNF- ⁇ ), and by improving other parameters which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer.
  • various cytokines e.g., IL-18, GM-CSF or IFN- ⁇
  • various cytokines e.g., IL-2, IL-4, IL-5, IL-10, or TNF- ⁇
  • a list of non-exhaustive examples of this includes extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • subject as used herein, is taken to mean any mammalian subject to which a human or bovine lactoferrin composition is orally, topical and/or parenterally 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 cancer.
  • genotypeic refers to cells, tissues or animals that have genotypes. For example, identical twins or animals of the same inbred strain. Syngeneic and isogeneic can be used interchangeable.
  • T-cell as used herein is defined as a thymus-derived cell that participates in a variety of cell-mediated immune reactions.
  • terapéuticaally effective amount refers to an amount that results in an improvement or remediation of the symptoms of the disease or condition.
  • treating and “treatment” as used herein 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.
  • vacuna as used herein is defined as material used to provoke an immune response (e.g., the production of antibodies) on administration of the materials and thus conferring immunity.
  • a vaccine is an antigenic and/or immunogenic composition.
  • 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, for example, U.S. Pat. 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 over natural LF and or rLF, e.g., the ability to stimulate and/or inhibit cytokines or chemokines.
  • 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.
  • deletional variants can be produced by proteolysis of lactoferrin and/or expression of a polynucleotide encoding a truncated lactoferrin as described in U.S. Pat. No. 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 tyrosine;
  • 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.
  • 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.
  • substitutional variants or replacement can be produced using standard mutagenesis techniques, for example, site-directed mutagenesis as disclosed in U.S. Pat. Nos. 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.
  • 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 N1 to N16 can be replaced or substituted.
  • a positively charged amino acid arginine, lysine, or histidine
  • a neutral amino acid alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylaline, proline, serine, th
  • 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, (e.g., IL-18, MIP-3 ⁇ , GM-CSF or IFN- ⁇ ) by inhibiting various cytokines, (e.g., IL-2, IL-4, IL-5, IL-10, and TNF- ⁇ ) and/or by improving the parameters related to which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer.
  • cytokines e.g., IL-18, MIP-3 ⁇ , GM-CSF or IFN- ⁇
  • cytokines e.g., IL-2, IL-4, IL-5, IL-10, and TNF- ⁇
  • N-terminal lactoferrin variants of the present invention are considered functional equivalents of lactoferrin.
  • biologically functional equivalents 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 lactoferrin to stimulate or inhibit various cytokines or chemokines and/or by improving the parameters which promotes or enhances which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer.
  • extension of the subject's life by any period of time decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • 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 derivatives and/or substitutions
  • lactoferrin composition a preparation of lactoferrin (lactoferrin composition)
  • 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
  • PTC-protein phenylthiocarbamyl derivative
  • 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.
  • the present invention is drawn to a composition comprising lactoferrin 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-1 terminal glycine residue is truncated or substituted.
  • N-terminal lactoferrin variants include variants that at least lack the N-terminal glycine residue or contain a substitution at the N-terminal glycine residue. The substitution can comprise substituting a natural or artificial amino acid residue for the N-terminal glycine residue.
  • the substitution can comprise substituting a positive amino acid residue or a negative amino acid residue for the N-terminal glycine residue or substituting a neutral amino acid residue other than glycine for the N-terminal glycine residue.
  • Other N-terminal lactoferrin variants include lactoferrin lacking one or more N-terminal residues or having one or more substitutions in the N-terminal.
  • 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.
  • the composition of the present invention suitable for administration is provided in a pharmaceutically acceptable carrier with or without an inert diluent.
  • the carrier should be assimilable and includes liquid, semi-solid, e.g., 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 the composition contained therein, its use in administrable composition 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, e.g., by solution, suspension, emulsification, admixture, encapsulation, absorption and the like. Such procedures are routine for those skilled in the art.
  • the composition 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, e.g., denaturation in the stomach.
  • stabilizers for use in an the composition include buffers, 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.
  • divalent metal chelators for example EDTA, can also be used to stabilize the composition of the present invention.
  • the stabilizer can also include antagonists to the secretion of stomach acids.
  • 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, parenteral, intramuscular, intraperitoneal, intravenous, intraarterial, intratumoral, topical or dermal.
  • Such compositions would normally be administered as pharmaceutically acceptable compositions as described herein.
  • 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, 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.
  • a powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • a liquid carrier such as, e.g., water or a saline solution
  • 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.
  • 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 0.01% and 5% wt/volume for application to the skin for treatment of cancer on or beneath the skin.
  • the amount of lactoferrin in the present invention may vary from about 1 mg to about 100 g of lactoferrin, more preferably 1 mg to 100 g per day.
  • the composition of the present invention comprises a lactoferrin concentration of about 0.01% to about 100%.
  • the lactoferrin composition may comprise lactoferrin or an N-terminal lactoferrin variant in which at least the N-1 terminal glycine residue is truncated and/or substituted.
  • 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 in combination with a cancer immunotherapy or vaccine to a subject suspected of or having cancer.
  • lactoferrin is acting an adjuvant.
  • the process involves administering a lactoferrin composition of the present invention and the immunotherapy 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 (lactoferrin and the cancer vaccine), 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 lactoferrin composition and the other includes the cancer vaccine. It is also contemplated that the lactoferrin composition and the cancer vaccine can be administered sequentially.
  • neoplasms include 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.
  • the neoplasm is a hematopoietic neoplasm which 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.
  • 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.
  • tumor-associated antigens have spurred the generation of new vaccine strategies (Sabel et al., 2002). These strategies can be divided into two categories: antigen-specific vaccines, in which the tumor antigens have been identified and can be isolated to develop a molecularly defined vaccine, and cellular or non-antigen specific, in which the tumor-specific antigens are unknown but presumed to exist within the material used to generate the vaccine (Borrello et al., 2002).
  • Tumor-specific antigens can be divided into four categories: unique tumor-specific antigens that are the products of mutation; viral antigens in virus-associated cancers; tissue-specific differentiation antigens; and tumor-selective antigens (D. Pardoll, 2002)
  • Active immunization against cancer can be achieved delivering tumor-specific peptide vaccines to patients along with an immune adjuvant meant to induce inflammation and stimulate immunity (Machiels et al., 2002).
  • An alternative approach is the use of cellular vaccines.
  • Autologous cellular vaccines present biologically relevant antigens to the immune system, but this is limited to individuals with sufficient tumor to prepare a vaccine (Chang et al., 2003)
  • Allogeneic cellular vaccines are based on the fact that tumor-associated antigens are shared among a large number of patients, so a vaccine prepared from a cultured cell line could stimulate an anti-tumor immune response in many patients (Vaishampayan et al., 2002).
  • ganglioside vaccines Knutson, 2002
  • viral oncolysates Horvath et al., 1999
  • anti-idiotype antibodies Alfonso et al., 2002
  • cytokine gene-modified tumor cell vaccines Formni et al., 2000
  • dendritic cell vaccines Curiel et al., 2002
  • DNA vaccines Bronte, 2001.
  • 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.
  • the present invention contemplates the treatment of cancer. It is envisioned that the present invention is directed at the use of the cancer immunotherapy in combination with a lactoferrin composition to treat subjects with cancer such that these subjects are conferred a therapeutic benefit as a result of the treatment.
  • a therapeutic benefit refers to a result that promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer.
  • a list of non-exhaustive examples of this includes extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • Treatment regimens may vary as well, and often depend on cancer vaccine used, 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.
  • lactoferrin is administered in an effective amount to potentiate the effect of the immunotherapy which is to decrease, reduce, inhibit or abrogate the growth of a tumor.
  • the amount of the lactoferrin composition may vary from about 1 mg to about 100 g of lactoferrin.
  • 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.
  • the present invention intends to provide, to a cell, an antigen-specific vaccines and/or DNA vaccines in combination with an effective amount of lactoferrin to potentiate the effect of the antigen-specific vaccine.
  • antigen-specific vaccines are incorporated into this section by reference.
  • one of skill in the art is well aware of how to apply gene delivery and/or DNA delivery to in vivo and ex vivo situations.
  • transduced antigen presenting cells and/or dendritic cell vaccines Another therapy that is contemplated is the administration of transduced antigen presenting cells and/or dendritic cell vaccines.
  • the antigen presenting cells and/or dendritic cells are transduced in vitro or ex vivo with an antigen-specific DNA.
  • the transduced antigen presenting cells and/or dendritic cells are then administered in combination with a lactoferrin composition.
  • the present invention includes a method of enhancing the immune response in an subject suffering from or susceptible to cancer comprising the steps of administering an antigen presenting cell that contains the DNA vaccine in combination with a lactoferrin composition.
  • the antigen presenting cells may be obtained from the blood of the subject or bone marrow of the subject. In certain preferred embodiments, the antigen presenting cells are isolated from the bone marrow. In a preferred embodiment, the antigen presenting cells are administered to the same or different subject (e.g., same or different donors). In a preferred embodiment, the subject has or is suspected of having a cancer,
  • a further embodiment of the present invention is a method of treating a cancer comprising the step of supplementing a mucosal or systemic immune system by increasing the amount of a lactoferrin composition in the gastrointestinal tract and/or in the systemic circulation.
  • 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 human lactoferrin.
  • human lactoferrin stimulates interleukin-18 in the gastrointestinal tract, which enhances immune cells.
  • interleukin-18 enhances T lymphocytes or natural killer cells.
  • interleukin-18 enhances CD4+, CD8+ and CD3+ cells. It is known by those of skill in the art that IL-18 is a Th1 cytokine that acts in synergy with interleukin-12 and interleukin-2 in the stimulation of lymphocyte IFN-gamma production.
  • cytokines may also be enhanced for example, but not limited to IL-1b or, IL-12 or IFN-gamma. It is also envisioned that human lactoferrin stimulates interleukin-18 following oral 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 cancer vaccine/adjuvant 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 cancer vaccine/adjuvant composition and the other includes the second agent(s).
  • the cancer vaccine/adjuvant 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 cancer vaccine/adjuvant 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 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), and Lomustine.
  • antibiotic chemotherapeutics such as
  • 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, 7-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 microscopically 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.
  • mice were challenged subcutaneously (s.c.) in the middle of the left flank with 0.2 ml of a single-cell suspension containing 1 ⁇ 10 5 TUBO cells.
  • Oral rhLF or placebo was administered (1000 mg/kg/once a day) two days before TUBO injection and for 3 weeks (five days/week with a total of 15 treatments). Tumors were measured twice a week for the duration of the experiment.
  • FIG. 1 shows that mice (4/group) treated with oral rhLF displayed a significant tumor inhibition (p ⁇ 0.05 vs. placebo or ctrl) whereas no activity was observed in mice treated with placebo or left untreated.
  • the adjuvant effect of oral LF in combination with DNA vaccination is evaluated. It is determined if oral LF in combination with a DNA vaccine will elicit complete protection against a lethal challenge of syngeneic carcinoma cells expressing p185 (TUBO) in BALB/C mice.
  • the DNA vaccine consists of a plasmid encoding for the transmembrane (TM) and extracellular domain (ECD) of the rat p185 (r-p185). Overexpression of p185 is frequent in human cancers and correlates with particular aggressiveness (Gullick et al., 1991).
  • the p185 vector is constructed as follows.
  • the pCMV vector is derived from the pcDNA3 plasmid (Invitrogen, San Diego, Calif.) by deleting the SV40 promoter, neomycin resistance gene, and SV40 poly(A).
  • the sequence for the ECD and that for the ECD and TM domain of mutated r-p185 are generated from the PCR product using the primers SEQ.ID.NO:1 3′-CGCAAGCTTCATCATGGAGCTGGC-5′ and SEQ.ID.NO:2 3′-CGGAATTCGGGCTGGCTCTCTGCTC-5′ and the primers SEQ.ID.NO:3 3′-CGCAAGCTTCATGGAGCTGGC-5′ and SEQ.ID.NO:4 3′-ATGAATTCTT TCCGCATCGTGTACTTCTTCCGG-5′, respectively, as described by Amici et al. (Cancer Immunol. Immunother., 1998).
  • PCR products of the expected size are isolated by agarose gel electrophoresis, digested with HindIII and EcoRI, and cloned into the multiple cloning site of the pCMV plasmid to obtain the two plasmids used in this work (ECD and ECD-TM plasmids).
  • the pCMVb (Clontech Laboratories, Palo Alto, Calif.) coding for ⁇ -galactosidase is used as a control plasmid ( ⁇ -gal plasmid).
  • Escherichia coli strain DH5a is transformed with ECD, ECD-TM, and ⁇ -gal plasmids and then grown in Luria-Bertani medium (Sigma, St.
  • mice are immunized twice intramuscularly (i.m.) with ECD-TM plasmid seven days before and after TUBO challenge with 14 days of interval between immunizations.
  • the plasmids (100 mg/injection) are injected into the quadriceps muscle through a 28-gauge needle syringe.
  • Oral lactoferrin (1000 mg/Kg) is administered daily starting seven days before TUBO challenge for three weeks of five treatments per week for a total 15 treatments.
  • Control animals are treated with oral placebo and DNA vaccination; no rhLF is administered to the control animals.
  • the efficacy of treatment is evaluated by measuring the solid tumor size during and at the end of the experiment. Tumor masses are measured bi-weekly with calipers in the two perpendicular diameters. Progressively growing masses of >3 mm in mean diameter are regarded as tumors. Growth is monitored until tumors exceed an average diameter of 10 mm, at which time mice are sacrificed for humane reasons.
  • the immune response is measured by the morphological analysis of tumor infiltration depicting CD4 + and CD8 + T lymphocytes, polymorhonuclear cells (PMN), macrophages, NK and dendritic cells. Expression of endothelial cell adhesion molecules is also analyzed in tumor vessels. Cytokine profiles obtained from tumors and immune cell infiltration provides further insight into the mechanism of action. Anti-r-p185 antibodies are also analyzed in the sera of ECD-TM vaccinated mice and treated with oral LF to detect an antibody response against TUBO. CTL activity against TUBO cells is also analyzed in spleen of mice treated with oral LF and ECD-TM immunized versus control groups.
  • the efficacy of treatment is evaluated by measuring the solid tumor size during and at the end of the experiment. Tumor masses are measured bi-weekly with calipers in the two perpendicular diameters. Progressively growing masses of >3 mm in mean diameter are regarded as tumors. Growth is monitored until tumors exceed an average diameter of 10 mm, at which time mice are sacrificed for humane reasons.
  • the immune response is measured by the morphological analysis of tumor infiltration depicting CD4 + and CD8 + T lymphocytes, polymorhonuclear cells (PMN), macrophages, NK and dendritic cells. Expression of endothelial cell adhesion molecules is also analyzed in tumor vessels. Cytokine profiles obtained from tumors and immune cell infiltration provides further insight into the mechanism of action. Anti-r-p185 antibodies are also analyzed in the sera of ECD-TM vaccinated mice and treated with oral LF to detect an antibody response against TUBO. CTL activity against TUBO cells is also analyzed in spleen of mice treated with oral LF and ECD-TM immunized versus control groups.
  • LF is orally administered daily for a three week course of five days/week followed by one week off from week 5 to 20 (total of five courses). Control mice receive oral placebo following the same schedule; no LF is administered to control animals.
  • mice Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters. Tumor-free mice (%) as well as mean number of tumors per mouse are recorded. Mice displaying progressively masses in all 10 mammary glands and those that reach 33 week of age are killed and morphologically examined.
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice.
  • the incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination.
  • Tumor progression is associated with neovascularization and lack of infiltrating CD8 + , and to a lesser extent CD4 + , lymphocytes evident by histology analysis of animals.
  • the level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • BALB-NeuT mice treated with oral LF are thymectomized at 4 week and receive 200 ug of anti-CD8 antibody intraperitoneally. Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters.
  • mice were immunized at the 14 th and 16 th week of age with the ECD-TM plasmid and treated with oral LF (1000 mg/Kg) daily for a three week course of five days/week followed by one week off from week 6 to 30 (total of seven courses) ( FIG. 3 ).
  • Control mice received only ECD-TM vaccination, oral FL, oral placebo (seven courses) or left untreated.
  • the efficacy of individual and combination treatments was evaluated by weekly inspection of mice to follow tumor onset and growth. Tumor masses were measured with calipers in the two perpendicular diameters and tumor-free mice (%) as well as mean number of tumors per mouse was recorded. Mice displaying progressively masses in all 10 mammary glands were sacrificed for humane reasons.
  • FIG. 4 shows that at week 34, when only 20% of mice vaccinated with ECD-TM did not have palpable mass in all 10 mammary glands, 80% of the mice treated with oral LF in combination with ECD-TM were tumor free ( FIG. 4A ). A significant reduction in tumor multiplicity was also evident ( FIG. 4B ).
  • Time of appearance of the first tumor ( FIG. 4A ) and mean number of palpable mammary carcinomas per mouse ( FIG. 4B ) in the group of 15 untreated (filled circle), 15 vaccinated (open circle), 5 placebo-treated (open square), 5 LF-treated (filled square) and 5 LF treated+vaccinated mice (filled diamond).
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice.
  • the incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination.
  • Tumor progression is associated with neovascularization and lack of infiltrating CD8 + , and to a lesser extent CD4 + , lymphocytes evident by histology analysis of animals.
  • the level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • BALB-NeuT mice immunized with ECD-TM plasmid and treated with oral LF are thymectomized at 4 week and receive 200 ug of anti-CD8 antibody intraperitoneally. Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters. Tumor-free mice (%) as well as mean number of tumors per mouse are recorded.
  • BALB-NeuT mice are vaccinated with allogeneic mammary carcinoma cells (Neu/H-2q) expressing high surface levels of both p185neu and H-2 q class I molecules (Nanni et al., 2001) and treated with oral LF. Beginning when mice are 6 week old, they receive Neu/H-2q cells twice-weekly in the first and second week followed by one week off. After one week of rest this course is repeated until week 33. Mice are also treated with oral LF (300 mg/Kg) daily for a three week course of five days/week followed by one week off from week 5 to 20 (total of five courses). Control mice receive oral placebo and allogeneic tumor cell vaccination schedule.
  • Neuronet al., 2001 allogeneic mammary carcinoma cells
  • mice The efficacy of individual and combination treatments is evaluated by weekly inspection of mice to follow tumor onset and growth. Tumor masses are measured with calipers in the two perpendicular diameters and tumor-free mice (%) as well as mean number of tumors per mouse is recorded. Mice displaying progressively masses in all 10 mammary glands and those that reach 33 week of age are killed and morphologically examined.
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice.
  • the incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination.
  • Tumor progression is associated with neovascularization and lack of infiltrating CD8 + , and to a lesser extent CD4 + , lymphocytes evident by histology analysis of animals.
  • the level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • rhLF is administered at a dose of 4.5 g per day (3 g in the morning, 1.5 g at night in cycles of 14 days on, 14 days off (maximum 5 cycles) or rhLF 9 g per day (4.5 g bid in cycles of 14 days on, 14 days off (maximum 5 cycles) to patients with metastatic HER-2/neu-overexpressing cancers.
  • the dose is administered orally.
  • Patients also receive monthly vaccinations with three 15-amino acid HER-2/neu-derived peptides containing within each the putative HLA-A2-binding motifs (Knutson et al., 2001). Five hundred micrograms of each peptide (1.5 microgram total peptide dose) are administered to the same draining lymph node site via two intradermal injections for 6 months.
  • 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 the mean peptide-specific T-cell precursor frequency to the HLA-A2 peptides. Peptide-specific T cell killing is also evaluated.
  • Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, and IL-4, IL-5, IL-10, IL-12 and IFN- ⁇ .
  • mice were administered live attenuated S typhimenium (Ty21a) by oral gavage at various doses. Animals also received oral rhLF (65 mg/kg) on the day before and on the day of the Ty21a administration. Stool samples were collected 19 days after Ty21a administration and mucosal immunity evaluated by measuring stool EspA specific sIgA antibodies. Results show that rhLF induces similar immunity with a 10,000 fold lower titre ( FIG. 2 ).

Abstract

The present invention relates to methods of treating cancer by administering a composition of lactoferrin (LF) in combination with cancer vaccines.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of prior application Ser. No. 10/862,213, filed Jun. 7, 2004, which claims the benefit of U.S. Provisional Application Nos. 60/476,318 filed Jun. 6, 2003 and 60/498,236 filed Aug. 27, 2003, all of which are incorporated herein by reference.
  • TECHNICAL FIELD
  • The present invention relates to methods of treating cancer by administering a composition of lactoferrin (LF) in combination with cancer vaccines.
  • BACKGROUND OF THE INVENTION
  • Currently, there are few effective options for the treatment of many common cancer types. The course of treatment for a given individual depends on the diagnosis, the stage to which the disease has developed, and factors such as age, sex, and general health of the patient. The most conventional options of cancer treatment are surgery, radiation therapy, and chemotherapy. Surgery plays a central role in the diagnosis and treatment of cancer. Typically, a surgical approach is required for biopsy and the removal of cancerous growth. However, if the cancer has metastasized and is widespread, surgery is unlikely to result in a cure, and an alternate approach must be taken. Side effects of surgery include diminished structural or organ function and increased risk of infection, bleeding, or coagulation related complications. Radiation therapy, 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).
  • Significant progress in understanding the molecular basis of the immune response to cancer as well as increased understanding of the basic mechanisms of cellular immunology have combined to open new opportunities for the development of effective immunotherapy for patients with cancer (Dudley et al., 2002). Immunotherapy includes both innate and specific immune responses that have the potential to treat different tumor types. The activation of tumor antigen-specific T lymphocytes or non-specific macrophages and natural killer (NK) cells using immunotherapeutic approaches may lead to the destruction of tumor cells (Curiel et al., 2002). Cancer vaccines involve the induction of a specific immune response. However, the administration of a tumor antigen alone is often not sufficient to stimulate an appropriate immune response. Incorporating an immunological adjuvant into a vaccine regimen often improves anti-tumor activity (Dredge et al., 2002).
  • 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.
  • Recombinant human lactoferrin has previously been described as being purified after expression in a variety of prokaryotic and eukaryotic organisms including aspergillus (U.S. Pat. No. 6,080,559), cattle (U.S. Pat. No. 5,919,913), rice, corn, Sacharomcyes (U.S. Pat. No. 6,228,614) and Pichia pastoris (U.S. Pat. No. 6,455,687, 6,277,817, 6,066,469). Also described are expression systems for the expression of full-length human lactoferrins (e.g., U.S. Pat. No. 6,100,054). In all cases, part of the teaching is expression of the full-length cDNA and purification of the intact protein whose N-terminal, after processing of the leader peptide, is the amino acid glycine. Nuijens et al. (U.S. Pat. No. 6,333,311) separately describe variants of human lactoferrin but their focus is limited to deletion or substitution of arginine residues found in the N-terminal domain of lactoferrin.
  • Recently, 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 cancer vaccine adjuvant for the prevention (prophylaxis) or treatment (therapeutic) of tumors.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention is directed to a method for preventing or treating cancer. The method of treatment involves administration of lactoferrin and a cancer immunotherapy, e.g., cancer vaccine. Thus, it is contemplated that lactoferrin can be used as an adjuvant for cancer immunotherapy.
  • The lactoferrin composition, which is dispersed in a pharmaceutically acceptable carrier, comprises lactoferrin or an N-terminal lactoferrin variant in which at least the N-terminal glycine residue is truncated or substituted. The lactoferrin is mammalian lactoferrin, more particularly, the lactoferrin is human or bovine. Yet further, the lactoferrin is recombinant lactoferrin. N-terminal lactoferrin variants include variants that at least lack the N-terminal glycine residue or contain a substitution at the N-terminal glycine residue. The substitution can comprise substituting a natural or artificial amino acid residue for the N-terminal glycine residue. For example, the substitution can comprise substituting a positive amino acid residue or a negative amino acid residue for the N-terminal glycine residue or substituting a neutral amino acid residue other than glycine for the N-terminal glycine residue. Other N-terminal lactoferrin variants include lactoferrin lacking one or more N-terminal residues or having one or more substitutions in the N-terminal. In specific embodiments, the 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 amount of the lactoferrin that is administered is about 1 mg to about 100 g per day, more preferably, the amount is about 10 mg to about 100 g per day. More particularly, the composition is a solution, capsule or a tablet having a lactoferrin concentration of about 0.01% to about 100%.
  • Another embodiment of the present invention comprises a method of treating cancer comprising the step of administering to a subject a cancer immunotherapy and an adjuvant, wherein the adjuvant is a lactoferrin composition that is administered in an amount sufficient to provide an improvement in the cancer in the subject. Still further, the method can further comprise additionally administering chemotherapy, immunotherapy, surgery, biotherapy, radiotherapy or a combination thereof to the subject.
  • It is envisioned that the lactoferrin composition can be administered orally, parenterally or topically. Parenterally includes, but is not limited subcutaneously, intramuscularly, intraperitoneally, intravenously, intraarterially, intramyocardially, transendocardially, transepicardially, or intrathecally. For oral administration, an antacid in combination with the lactoferrin composition can be administered. The lactoferrin composition can be formulated in a delayed release formulation. Still further, the lactoferrin composition can be formulated wherein release occurs in the small intestine or in the large intestine.
  • In specific embodiments, the cancer 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. More particularly, the neoplasm is a hematopoietic neoplasm. Exemplary hematopoietic neoplasms include, but are not limited to myelogenous leukemia, acute lymphoblastic leukemia, myelodysplastic syndrome, chronic myelomonocytic leukemia, juvenile myelomonocyte leukemia, multiple myeloma, and chronic lymphocytic leukemia.
  • In certain embodiments, the immunotherapy comprises antigen presenting cells. More specifically, the lactoferrin composition is administered ex vivo to the antigen presenting cells prior to administering the cells to the subject. The cells are allogeneic or syngeneic.
  • Still further, the immunotherapy comprises administration of a tumor antigen to the subject, or administration of a nucleic acid sequence expressing a cancer antigen to the subject. In specific embodiments, the nucleic acid sequence is contained in a vector.
  • Yet further, the immunotherapy comprises administration of a vector containing a nucleic acid sequence expressing an immunomodulatory cytokine to the subject.
  • Still further, the immunotherapy comprises administration of a protein or nucleic acid that promotes the recognition of a cancer antigen in the subject.
  • In certain embodiments, the lactoferrin composition is administered simultaneously and/or sequentially with the immunotherapy.
  • Another method of the present invention comprises a method of enhancing the immune system in a subject suffering from cancer or susceptible to cancer comprising the step of administering to the subject a cancer immunotherapy and an adjuvant, wherein the adjuvant is a lactoferrin composition. More specifically, the lactoferrin is administered orally.
  • In certain embodiments, lactoferrin stimulates the production of interleukin-18, GM-CSF, or MIP-3alpha. Yet further, interleukin-18, GM-CSF or MIP-3alpha stimulate the production, maturation, migration or activity of immune cells (e.g., T lymphocytes, natural killer cells, dendritic cells, antigen presenting cells or progenitor cell). More specifically, T lymphocytes are selected from the group consisting of CD4+, CD8+ and CD3+ cells.
  • 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 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 that such equivalent constructions do not depart from 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
  • 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:
  • FIG. 1 shows carcinoma cell tumor growth with and without oral administration of lactoferrin.
  • FIG. 2 shows the effect of rhLF on mucosal immunity in mice.
  • FIG. 3 shows and ECD-TM vaccination and oral LF schedule.
  • FIG. 4A and FIG. 4B show the effect of oral LF in combination with ECD-TM vaccination n carcinogenesis in BALB-neuT mice.
  • DETAILED DESCRIPTION OF THE INVENTION
  • 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.
  • I. DEFINITIONS
  • 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.
  • The term “adjuvant” as used herein refers to substance added to drug product formulation that affects the action of the active ingredient by enhancing or potentiating its activity. An adjuvant also includes a treatment and/or therapy that is added or combined with a traditional treatment and/or therapy to enhance or potentiate or extend the effect of the traditional treatment and/or therapy.
  • The term “allogeneic” as used herein, refers to cell types or tissues that are antigenically distinct. Thus, cells or tissue transferred from the same species can be antigenically distinct.
  • The term “antigen” as used herein is defined as a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both. An antigen can be derived from organisms, subunits of proteins/antigens, killed or inactivated whole cells or lysates. Therefore, a skilled artisan realizes that any macromolecule, including virtually all proteins or peptides, can serve as antigens. Furthermore, antigens can be derived from recombinant or genomic DNA. A skilled artisan realizes that any DNA, which contains nucleotide sequences or partial nucleotide sequences of a pathogenic genome or a gene or a fragment of a gene for a protein that elicits an immune response results in synthesis of an antigen.
  • The term “antigen presenting cell” as used herein is any cell that enhances the immune response (i.e., from the T-cell or —B-cell arms of the immune system) against an antigen or antigenic composition.
  • The term “dendritic cell” or “DC” as used herein is defined as an example of an antigen presenting cell derived from bone marrow. Dendritic cells have a branched or dendritic morphology and are the most potent stimulations of T-cell response.
  • As used herein, the term “ex vivo” refers to “outside” the body. One of skill in the art is aware that ex vivo and in vitro can be used interchangeably.
  • 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, intratumoral, intraocular, or intraarticular administration.
  • The term “oral administration” as used herein includes oral, buccal, enteral or intragastric administration.
  • The term “topical administration” as used herein includes application to a dermal, epidermal, subcutaneous or mucosal surface.
  • 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 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.
  • The term “lactoferrin composition” as used herein refers to a composition having lactoferrin, a portion or part of lactoferrin, an N-terminal lactoferrin variant, or a combination thereof.
  • The term “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.
  • The term “human lactoferrin” or “hLF” as used herein refers to native or recombinant human lactoferrin. Native human lactoferrin can be obtained by purification from human milk or colostrum or from other natural sources. Recombinant human lactoferrin (rhLF) 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.
  • The term “bovine lactoferrin” or “bLF’ as used herein refers to native or recombinant bovine lactoferrin. Native bovine lactoferrin can be obtained by purification from bovine milk. Recombinant bovine lactoferrin (rbLF) 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.
  • 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 (e.g., IL-18, GM-CSF or IFN-γ) by inhibiting various cytokines, (e.g., IL-2, IL-4, IL-5, IL-10, or TNF-α), and by improving other parameters which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer. A list of non-exhaustive examples of this includes extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • The term “subject” as used herein, is taken to mean any mammalian subject to which a human or bovine lactoferrin composition is orally, topical and/or parenterally 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 cancer.
  • As used herein, the term “syngeneic” refers to cells, tissues or animals that have genotypes. For example, identical twins or animals of the same inbred strain. Syngeneic and isogeneic can be used interchangeable.
  • The term “T-cell” as used herein is defined as a thymus-derived cell that participates in a variety of cell-mediated immune reactions.
  • 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.
  • The term “treating” and “treatment” as used herein 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. 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.
  • The term “vaccine” as used herein is defined as material used to provoke an immune response (e.g., the production of antibodies) on administration of the materials and thus conferring immunity. Thus, a vaccine is an antigenic and/or immunogenic composition.
  • II. LACTOFERRIN
  • 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, for example, U.S. Pat. Nos. 5,571,896; 5,571,697 and 5,571,691, which are herein incorporated by reference.
  • In certain aspects, the present invention provides lactoferrin variants having enhanced biological activities over natural LF and or rLF, e.g., the ability to stimulate and/or inhibit cytokines or chemokines. 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.
  • The deletional variants can be produced by proteolysis of lactoferrin and/or expression of a polynucleotide encoding a truncated lactoferrin as described in U.S. Pat. No. 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 tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine.
  • 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.
  • 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).
  • 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.
  • 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. Pat. No. 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. Pat. No. 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).
  • 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.
  • Thus, in the present invention, substitutional variants or replacement can be produced using standard mutagenesis techniques, for example, site-directed mutagenesis as disclosed in U.S. Pat. Nos. 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. It is envisioned that 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 N1 to N16 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, (e.g., IL-18, MIP-3α, GM-CSF or IFN-γ) by inhibiting various cytokines, (e.g., IL-2, IL-4, IL-5, IL-10, and TNF-α) and/or by improving the parameters related to which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer. A list of non-exhaustive examples of this includes extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition. Thus, the N-terminal lactoferrin variants of the present invention are considered functional equivalents of lactoferrin.
  • 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 lactoferrin to stimulate or inhibit various cytokines or chemokines and/or by improving the parameters which promotes or enhances which promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer. For example, extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • 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.
  • TABLE 1
    Modified and/or Unusual Amino Acids
    Abbr. Amino Acid Abbr. Amino Acid
    Aad 2-Aminoadipic acid EtAsn N-Ethylasparagine
    BAad 3-Aminoadipic acid Hyl Hydroxylysine
    BAla beta-alanine, beta-Amino-propionic AHyl allo-Hydroxylysine
    acid
    Abu 2-Aminobutyric acid 3Hyp 3-Hydroxyproline
    4Abu 4-Aminobutyric acid, piperidinic 4Hyp 4-Hydroxyproline
    acid
    Acp 6-Aminocaproic acid Ide Isodesmosine
    Ahe 2-Aminoheptanoic acid Aile allo-Isoleucine
    Aib 2-Aminoisobutyric acid MeGly N-Methylglycine,
    sarcosine
    BAib 3-Aminoisobutyric acid MeIle N-Methylisoleucine
    Apm 2-Aminopimelic acid MeLys 6-N-Methyllysine
    Dbu
    2,4-Diaminobutyric acid MeVal N-Methylvaline
    Des Desmosine Nva Norvaline
    Dpm
    2,2′-Diaminopimelic acid Nle Norleucine
    Dpr
    2,3-Diaminopropionic acid Orn Ornithine
    EtGly N-Ethylglycine
  • 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.
  • 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.
  • 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.
  • III. PHARMACEUTICAL COMPOSITIONS
  • The present invention is drawn to a composition comprising lactoferrin 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-1 terminal glycine residue is truncated or substituted. N-terminal lactoferrin variants include variants that at least lack the N-terminal glycine residue or contain a substitution at the N-terminal glycine residue. The substitution can comprise substituting a natural or artificial amino acid residue for the N-terminal glycine residue. For example, the substitution can comprise substituting a positive amino acid residue or a negative amino acid residue for the N-terminal glycine residue or substituting a neutral amino acid residue other than glycine for the N-terminal glycine residue. Other N-terminal lactoferrin variants include lactoferrin lacking one or more N-terminal residues or having one or more substitutions in the N-terminal. 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.
  • Further in accordance with the present invention, the composition of the present invention suitable for administration is provided in a pharmaceutically acceptable carrier with or without an inert diluent. The carrier should be assimilable and includes liquid, semi-solid, e.g., 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 the composition contained therein, its use in administrable composition for use in practicing the methods of the present invention is appropriate. Examples of carriers or diluents include fats, oils, water, saline solutions, lipids, liposomes, resins, binders, fillers and the like, or combinations thereof.
  • In accordance with the present invention, the composition is combined with the carrier in any convenient and practical manner, e.g., by solution, suspension, emulsification, admixture, encapsulation, absorption and the like. Such procedures are routine for those skilled in the art.
  • In a specific embodiment of the present invention, the composition 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, e.g., denaturation in the stomach. Examples of stabilizers for use in an the composition include buffers, 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. Yet further, it is envisioned that divalent metal chelators, for example EDTA, can also be used to stabilize the composition of the present invention. More preferably, for an orally administered composition, the stabilizer can also include antagonists to the secretion of stomach acids.
  • Administration of the 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, parenteral, intramuscular, intraperitoneal, intravenous, intraarterial, intratumoral, topical or dermal. Such compositions would normally be administered as pharmaceutically acceptable compositions as described herein.
  • The 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, 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.
  • In another embodiment, a powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • The 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. Generally, 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. In the case of sterile powders for the preparation of sterile injectable solutions, 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.
  • Further, 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. Specifically, a powdered lactoferrin composition is combined with an aqueous gel containing an polymerization agent such as Carbopol 980 at strengths between 0.01% and 5% wt/volume for application to the skin for treatment of cancer on or beneath the skin.
  • The amount of lactoferrin in the present invention may vary from about 1 mg to about 100 g of lactoferrin, more preferably 1 mg to 100 g per day. In preferred embodiments, the composition of the present invention comprises a lactoferrin concentration of about 0.01% to about 100%. The lactoferrin composition may comprise lactoferrin or an N-terminal lactoferrin variant in which at least the N-1 terminal glycine residue is truncated and/or substituted.
  • 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 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.
  • IV. TREATMENT OF CANCER
  • In accordance with the present invention, a lactoferrin composition provided in any of the above-described pharmaceutical carriers is administered in combination with a cancer immunotherapy or vaccine to a subject suspected of or having cancer. Thus, one of skill in the art realizes that lactoferrin is acting an adjuvant. The process involves administering a lactoferrin composition of the present invention and the immunotherapy 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 (lactoferrin and the cancer vaccine), 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 lactoferrin composition and the other includes the cancer vaccine. It is also contemplated that the lactoferrin composition and the cancer vaccine can be administered sequentially.
  • Cancer, 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.
  • More particularly, the neoplasm is a hematopoietic neoplasm which 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.
  • A. Immunotherapy
  • 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. 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.
  • Improvements in the identification of tumor-associated antigens have spurred the generation of new vaccine strategies (Sabel et al., 2002). These strategies can be divided into two categories: antigen-specific vaccines, in which the tumor antigens have been identified and can be isolated to develop a molecularly defined vaccine, and cellular or non-antigen specific, in which the tumor-specific antigens are unknown but presumed to exist within the material used to generate the vaccine (Borrello et al., 2002). Tumor-specific antigens can be divided into four categories: unique tumor-specific antigens that are the products of mutation; viral antigens in virus-associated cancers; tissue-specific differentiation antigens; and tumor-selective antigens (D. Pardoll, 2002)
  • Active immunization against cancer can be achieved delivering tumor-specific peptide vaccines to patients along with an immune adjuvant meant to induce inflammation and stimulate immunity (Machiels et al., 2002). An alternative approach is the use of cellular vaccines. Autologous cellular vaccines present biologically relevant antigens to the immune system, but this is limited to individuals with sufficient tumor to prepare a vaccine (Chang et al., 2003) Allogeneic cellular vaccines are based on the fact that tumor-associated antigens are shared among a large number of patients, so a vaccine prepared from a cultured cell line could stimulate an anti-tumor immune response in many patients (Vaishampayan et al., 2002). Several additional approaches to vaccine therapies include among others ganglioside vaccines (Knutson, 2002) viral oncolysates (Horvath et al., 1999), anti-idiotype antibodies (Alfonso et al., 2002), cytokine gene-modified tumor cell vaccines (Forni et al., 2000), dendritic cell vaccines (Curiel et al., 2002), and DNA vaccines (Bronte, 2001).
  • 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.
  • B. Treatments
  • The present invention contemplates the treatment of cancer. It is envisioned that the present invention is directed at the use of the cancer immunotherapy in combination with a lactoferrin composition to treat subjects with cancer such that these subjects are conferred a therapeutic benefit as a result of the treatment. Thus, a therapeutic benefit refers to a result that promotes or enhances the well-being of the subject with respect to the medical treatment of his/her cancer. A list of non-exhaustive examples of this includes extension of the subject's life by any period of time; decrease or delay in the neoplastic development of the disease; decrease in hyperproliferation; reduction in tumor growth; delay of metastases; reduction in the proliferation rate of a cancer cell, tumor cell, or any other hyperproliferative cell; induction of apoptosis in any treated cell or in any cell affected by a treated cell; and a decrease in pain to the subject that can be attributed to the subject's condition.
  • Treatment regimens may vary as well, and often depend on cancer vaccine used, 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.
  • In a preferred embodiment of the present invention, lactoferrin is administered in an effective amount to potentiate the effect of the immunotherapy which is to decrease, reduce, inhibit or abrogate the growth of a tumor. The amount of the lactoferrin composition may vary from about 1 mg to about 100 g of lactoferrin.
  • In specific embodiments, 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. In a further embodiment, 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.
  • Specifically, the present invention intends to provide, to a cell, an antigen-specific vaccines and/or DNA vaccines in combination with an effective amount of lactoferrin to potentiate the effect of the antigen-specific vaccine. The discussion of antigen-specific vaccines is incorporated into this section by reference. Thus, one of skill in the art is well aware of how to apply gene delivery and/or DNA delivery to in vivo and ex vivo situations.
  • Another therapy that is contemplated is the administration of transduced antigen presenting cells and/or dendritic cell vaccines. The antigen presenting cells and/or dendritic cells are transduced in vitro or ex vivo with an antigen-specific DNA. The transduced antigen presenting cells and/or dendritic cells are then administered in combination with a lactoferrin composition.
  • Still further, the present invention includes a method of enhancing the immune response in an subject suffering from or susceptible to cancer comprising the steps of administering an antigen presenting cell that contains the DNA vaccine in combination with a lactoferrin composition. The antigen presenting cells may be obtained from the blood of the subject or bone marrow of the subject. In certain preferred embodiments, the antigen presenting cells are isolated from the bone marrow. In a preferred embodiment, the antigen presenting cells are administered to the same or different subject (e.g., same or different donors). In a preferred embodiment, the subject has or is suspected of having a cancer,
  • A further embodiment of the present invention is a method of treating a cancer comprising the step of supplementing a mucosal or systemic immune system by increasing the amount of a lactoferrin composition in the gastrointestinal tract and/or in the systemic circulation.
  • 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 human lactoferrin. It is envisioned that human lactoferrin stimulates interleukin-18 in the gastrointestinal tract, which enhances immune cells. For example, interleukin-18 enhances T lymphocytes or natural killer cells. In specific embodiments, interleukin-18 (IL-18) enhances CD4+, CD8+ and CD3+ cells. It is known by those of skill in the art that 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. It is also envisioned that human lactoferrin stimulates interleukin-18 following oral administration, which inhibits angiogenesis and thereby has activity against tumor cells which are dependent on neovascularization.
  • V. COMBINATION TREATMENTS
  • In order to increase the effectiveness of the cancer vaccine/adjuvant (lactoferrin composition of the present invention), it may be desirable to combine the cancer vaccine/adjuvant composition of the present invention with other agents effective in the treatment of cancer, 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 cancer vaccine/adjuvant 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 cancer vaccine/adjuvant composition and the other includes the second agent(s).
  • Alternatively, the cancer vaccine/adjuvant 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 cancer vaccine/adjuvant 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
  • 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 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), and Lomustine.
  • 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.
  • B. Radiotherapeutic Agents
  • Radiotherapeutic agents and factors include radiation and waves that induce DNA damage for example, 7-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.
  • C. Surgery
  • 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.
  • 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 microscopically 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.
  • 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
  • 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.
  • 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. 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.
  • 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. 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-1, MIP-1beta, MCP-1, RANTES, and other chemokines.
  • VI. EXAMPLES
  • 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 Oral LF Inhibits the Growth of Her-2/neu+ Transplantable Carcinoma (TUBO)
  • Briefly, BALB/C mice were challenged subcutaneously (s.c.) in the middle of the left flank with 0.2 ml of a single-cell suspension containing 1×105 TUBO cells. Oral rhLF or placebo was administered (1000 mg/kg/once a day) two days before TUBO injection and for 3 weeks (five days/week with a total of 15 treatments). Tumors were measured twice a week for the duration of the experiment. FIG. 1 shows that mice (4/group) treated with oral rhLF displayed a significant tumor inhibition (p<0.05 vs. placebo or ctrl) whereas no activity was observed in mice treated with placebo or left untreated.
  • Results from this study show that rhLF administered orally significantly (p<0.05) inhibits tumor growth of a transplantable carcinoma over-expressing the Her-2/neu (r-p185) oncogene. Based upon these results, it is further contemplated that oral lactoferrin affects the tumor by enhancing immune cell activity against tumor-specific antigen (Her-2/neu).
  • Example 2 Evaluation of rhLF as Adjuvant of p185 DNA Vaccine in the Prevention of TUBO
  • The adjuvant effect of oral LF in combination with DNA vaccination is evaluated. It is determined if oral LF in combination with a DNA vaccine will elicit complete protection against a lethal challenge of syngeneic carcinoma cells expressing p185 (TUBO) in BALB/C mice. The DNA vaccine consists of a plasmid encoding for the transmembrane (TM) and extracellular domain (ECD) of the rat p185 (r-p185). Overexpression of p185 is frequent in human cancers and correlates with particular aggressiveness (Gullick et al., 1991).
  • The p185 vector is constructed as follows. The pCMV vector is derived from the pcDNA3 plasmid (Invitrogen, San Diego, Calif.) by deleting the SV40 promoter, neomycin resistance gene, and SV40 poly(A). The sequence for the ECD and that for the ECD and TM domain of mutated r-p185 are generated from the PCR product using the primers SEQ.ID.NO:1 3′-CGCAAGCTTCATCATGGAGCTGGC-5′ and SEQ.ID.NO:2 3′-CGGAATTCGGGCTGGCTCTCTGCTC-5′ and the primers SEQ.ID.NO:3 3′-CGCAAGCTTCATGGAGCTGGC-5′ and SEQ.ID.NO:4 3′-ATGAATTCTT TCCGCATCGTGTACTTCTTCCGG-5′, respectively, as described by Amici et al. (Cancer Immunol. Immunother., 1998). PCR products of the expected size are isolated by agarose gel electrophoresis, digested with HindIII and EcoRI, and cloned into the multiple cloning site of the pCMV plasmid to obtain the two plasmids used in this work (ECD and ECD-TM plasmids). The pCMVb (Clontech Laboratories, Palo Alto, Calif.) coding for β-galactosidase is used as a control plasmid (β-gal plasmid). Escherichia coli strain DH5a is transformed with ECD, ECD-TM, and β-gal plasmids and then grown in Luria-Bertani medium (Sigma, St. Louis, Mo.). Large-scale preparation of the plasmids is conducted by alkaline lysis using Endofree Qiagen Plasmid-Giga kits (Qiagen, Chatsworth, Calif.). DNA is then precipitated, suspended in sterile saline at the concentration of 1 mg/ml, and stored in aliquots at −20° C. for subsequent use in immunization protocols.
  • BALB/C mice are immunized twice intramuscularly (i.m.) with ECD-TM plasmid seven days before and after TUBO challenge with 14 days of interval between immunizations. The plasmids (100 mg/injection) are injected into the quadriceps muscle through a 28-gauge needle syringe. Oral lactoferrin (1000 mg/Kg) is administered daily starting seven days before TUBO challenge for three weeks of five treatments per week for a total 15 treatments. Control animals are treated with oral placebo and DNA vaccination; no rhLF is administered to the control animals.
  • The efficacy of treatment is evaluated by measuring the solid tumor size during and at the end of the experiment. Tumor masses are measured bi-weekly with calipers in the two perpendicular diameters. Progressively growing masses of >3 mm in mean diameter are regarded as tumors. Growth is monitored until tumors exceed an average diameter of 10 mm, at which time mice are sacrificed for humane reasons.
  • The immune response is measured by the morphological analysis of tumor infiltration depicting CD4+ and CD8+ T lymphocytes, polymorhonuclear cells (PMN), macrophages, NK and dendritic cells. Expression of endothelial cell adhesion molecules is also analyzed in tumor vessels. Cytokine profiles obtained from tumors and immune cell infiltration provides further insight into the mechanism of action. Anti-r-p185 antibodies are also analyzed in the sera of ECD-TM vaccinated mice and treated with oral LF to detect an antibody response against TUBO. CTL activity against TUBO cells is also analyzed in spleen of mice treated with oral LF and ECD-TM immunized versus control groups.
  • Example 3 Evaluation of rhLF as Adjuvant of p185 DNA Vaccine in the Treatment of TUBO
  • In this study, the adjuvant effect of oral LF in combination with DNA vaccination to inhibit the growth of established carcinoma expressing p185 (TUBO) in BALB/C mice is evaluated.
  • The efficacy of treatment is evaluated by measuring the solid tumor size during and at the end of the experiment. Tumor masses are measured bi-weekly with calipers in the two perpendicular diameters. Progressively growing masses of >3 mm in mean diameter are regarded as tumors. Growth is monitored until tumors exceed an average diameter of 10 mm, at which time mice are sacrificed for humane reasons.
  • The immune response is measured by the morphological analysis of tumor infiltration depicting CD4+ and CD8+ T lymphocytes, polymorhonuclear cells (PMN), macrophages, NK and dendritic cells. Expression of endothelial cell adhesion molecules is also analyzed in tumor vessels. Cytokine profiles obtained from tumors and immune cell infiltration provides further insight into the mechanism of action. Anti-r-p185 antibodies are also analyzed in the sera of ECD-TM vaccinated mice and treated with oral LF to detect an antibody response against TUBO. CTL activity against TUBO cells is also analyzed in spleen of mice treated with oral LF and ECD-TM immunized versus control groups.
  • Example 4 Oral LF in the Inhibition of Spontaneous Carcinomas
  • LF is orally administered daily for a three week course of five days/week followed by one week off from week 5 to 20 (total of five courses). Control mice receive oral placebo following the same schedule; no LF is administered to control animals.
  • Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters. Tumor-free mice (%) as well as mean number of tumors per mouse are recorded. Mice displaying progressively masses in all 10 mammary glands and those that reach 33 week of age are killed and morphologically examined.
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice. The incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination. Tumor progression is associated with neovascularization and lack of infiltrating CD8+, and to a lesser extent CD4+, lymphocytes evident by histology analysis of animals. The level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • To evaluate the role of CD8+ T cells in response to oral LF, BALB-NeuT mice treated with oral LF (same schedule as above) are thymectomized at 4 week and receive 200 ug of anti-CD8 antibody intraperitoneally. Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters.
  • Example 5 Oral LF and DNA Vaccination in the Inhibition of Spontaneous Carcinomas
  • In this study, the ability of oral LF in combination with DNA vaccination to hamper the aggressive carcinogenesis that takes place in all mammary glands of BALB-NeuT mice is evaluated.
  • BALB-NeuT mice were immunized at the 14th and 16th week of age with the ECD-TM plasmid and treated with oral LF (1000 mg/Kg) daily for a three week course of five days/week followed by one week off from week 6 to 30 (total of seven courses) (FIG. 3). Control mice received only ECD-TM vaccination, oral FL, oral placebo (seven courses) or left untreated. The efficacy of individual and combination treatments was evaluated by weekly inspection of mice to follow tumor onset and growth. Tumor masses were measured with calipers in the two perpendicular diameters and tumor-free mice (%) as well as mean number of tumors per mouse was recorded. Mice displaying progressively masses in all 10 mammary glands were sacrificed for humane reasons.
  • FIG. 4 shows that at week 34, when only 20% of mice vaccinated with ECD-TM did not have palpable mass in all 10 mammary glands, 80% of the mice treated with oral LF in combination with ECD-TM were tumor free (FIG. 4A). A significant reduction in tumor multiplicity was also evident (FIG. 4B).
  • Time of appearance of the first tumor (FIG. 4A) and mean number of palpable mammary carcinomas per mouse (FIG. 4B) in the group of 15 untreated (filled circle), 15 vaccinated (open circle), 5 placebo-treated (open square), 5 LF-treated (filled square) and 5 LF treated+vaccinated mice (filled diamond).
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice. The incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination. Tumor progression is associated with neovascularization and lack of infiltrating CD8+, and to a lesser extent CD4+, lymphocytes evident by histology analysis of animals. The level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • To evaluate the role of CD8+ T cells in response to oral LF, and DNA vaccination, BALB-NeuT mice immunized with ECD-TM plasmid and treated with oral LF (same schedule as above) are thymectomized at 4 week and receive 200 ug of anti-CD8 antibody intraperitoneally. Starting at the age of five week, mammary glands of each mouse are inspected once a week, and masses are measured with calipers in the two perpendicular diameters. Tumor-free mice (%) as well as mean number of tumors per mouse are recorded.
  • Example 6 Oral LF in Combination with Tumor Cell Vaccination in the Inhibition of Spontaneous Carcinoma
  • The ability of oral LF in combination with allogeneic tumor vaccination to hamper mammary carcinogenesis in HER-2/neu transgenic mice is evaluated.
  • BALB-NeuT mice are vaccinated with allogeneic mammary carcinoma cells (Neu/H-2q) expressing high surface levels of both p185neu and H-2q class I molecules (Nanni et al., 2001) and treated with oral LF. Beginning when mice are 6 week old, they receive Neu/H-2q cells twice-weekly in the first and second week followed by one week off. After one week of rest this course is repeated until week 33. Mice are also treated with oral LF (300 mg/Kg) daily for a three week course of five days/week followed by one week off from week 5 to 20 (total of five courses). Control mice receive oral placebo and allogeneic tumor cell vaccination schedule. The efficacy of individual and combination treatments is evaluated by weekly inspection of mice to follow tumor onset and growth. Tumor masses are measured with calipers in the two perpendicular diameters and tumor-free mice (%) as well as mean number of tumors per mouse is recorded. Mice displaying progressively masses in all 10 mammary glands and those that reach 33 week of age are killed and morphologically examined.
  • Histological examination of the mammary gland at progressive time points in a fashion blind to the treatment reveals the progression of the HER-2/neu oncogenesis in the treated mice. The incidence of atypical hyperplasia (week 10) and the number of carcinoma in situ (week 15) shows the anti-tumor activity induced by oral LF in combination with DNA vaccination. Tumor progression is associated with neovascularization and lack of infiltrating CD8+, and to a lesser extent CD4+, lymphocytes evident by histology analysis of animals. The level of vascularization and T cell infiltration in tumors treated with LF reveals the activity of oral LF.
  • Example 7 Oral Administration of hLF in Combination with a Cancer Vaccine in Patients
  • Recombinant human lactoferrin is orally administered to human patients in combination with a cancer vaccine to inhibit tumor growth. The cancer vaccine is the HER-2/neu. The following peptides used in this study are HER-2/neu peptides, p369-384, SEQ.ID.NO:5 KIFGSLAFLPESFDGDPA (Disis et al., 2000), p688-703, SEQ.ID.NO:6 RRLLQETELVEPLTPS (Disis et al., 2000), p971-984, SEQ.ID.NO:7 ELVSEFSRMARDPQ (Disis et al., 2000), p369-377, SEQ.ID.NO:8 KIFGSLAFL (Fisk et al., 1995), p689-697, SEQ.ID.NO:9 RLLQETELV (Peoples et al., 1995), and p971-979, SEQ.ID.NO:10 ELVSEFSRM (Ioannides et al., 1993).
  • Briefly, rhLF is administered at a dose of 4.5 g per day (3 g in the morning, 1.5 g at night in cycles of 14 days on, 14 days off (maximum 5 cycles) or rhLF 9 g per day (4.5 g bid in cycles of 14 days on, 14 days off (maximum 5 cycles) to patients with metastatic HER-2/neu-overexpressing cancers. The dose is administered orally.
  • Patients also receive monthly vaccinations with three 15-amino acid HER-2/neu-derived peptides containing within each the putative HLA-A2-binding motifs (Knutson et al., 2001). Five hundred micrograms of each peptide (1.5 microgram total peptide dose) are administered to the same draining lymph node site via two intradermal injections for 6 months.
  • 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 the mean peptide-specific T-cell precursor frequency to the HLA-A2 peptides. Peptide-specific T cell killing is also evaluated. Plasma, serum and blood cell extract samples are collected to measure circulating IL-18, IL-1, IL-2, and IL-4, IL-5, IL-10, IL-12 and IFN-γ.
  • Example 8 Effect of rhLF on Mucosal Immunity in Mice
  • BALBc mice were administered live attenuated S typhimenium (Ty21a) by oral gavage at various doses. Animals also received oral rhLF (65 mg/kg) on the day before and on the day of the Ty21a administration. Stool samples were collected 19 days after Ty21a administration and mucosal immunity evaluated by measuring stool EspA specific sIgA antibodies. Results show that rhLF induces similar immunity with a 10,000 fold lower titre (FIG. 2).
  • REFERENCES CITED
  • 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. Pat. No. 5,629,001
    • Alfonso M, et al., J Immunol. 168:2523-9, 2002.
    • Amici et al., Cancer Immunol. Immunother., 47:183-90. 1998.
    • Bezault J et. al., Cancer Res. 1994, 54(9):2310-2.
    • Boggio et al., J. Exp. Med., 1998, 188:589-596
    • Borrello I M, et al., Cancer Control 2002, 9:138-151
    • Bronte V, Curr. Gene Ther. 2001, 1:53-100
    • Broxmeyer H E. Blood. 1983; 61:982-993.
    • Chang A E, et al., J. Clin. Oncol. 21:884-90
    • Curiel T, et al. J. Clin. Invest. 2002, 109:311-312.
    • Damiens E, et al., Biochim Biophys Acta. 1998, 1402(3):277-87.
    • Dhennin-Duthille I, et al., J Cell Biochem. 2000, 79(4):583-93.
    • Disis et al. Transplantation, 2000.
    • Disis et al., Breast Cancer Res Treat, 62:245-52, 2000.
    • Dredge K, et al., Cancer Immunol. Immunother. 2002, 51(10):521-31
    • Dudley M et al., Science. 2002, 298(5594):850-4
    • Erlandsson, Cancer Genet. Cytogenet, 104:1-18, 1998.
    • Fisk et al., J. Exp. Med., 181:2109-17, 1995.
    • Forni G, et al. Curr. Opin. Mol. Ther. 1999, 1:34-8
    • Gahr M, et al., J Leukocyte Biol. 1991; 49: 427-33.
    • Gertig and Hunter, Semin. Cancer Biol., 8(4):285-298, 1997.
    • Gullick et al., Br. 1991, J. Cancer, 63:434.
    • Horowitz D A, et al., J Immunol. 1984; 132: 2370-4.
    • Horvath et al., Acta Microbiol Immunol. Hung, 1999, 46:1-20
    • Iigo M, et al., Clin Exp Metastasis. 1999, 17(1):35-40.
    • Ioannides et al., Cell. Immunol., 151:225-34, 1993.
    • Knutson K L et al., J. Clin. Invest., 2001, 107:477.
    • Knutson K L, Curr. Opin. Investig. Drugs 2002, 3:159-64
    • Kolmel, J. Neurooncol., 38:121-125, 1998.
    • Kuhara T, et al., Nutr Cancer. 2000, 38(2):192-9.
    • Machiels J P, et al., 2002, 29:459-502
    • Magi-Galluzzi et al., Anal. Quant. Cytol. Histol., 20:343-350, 1998.
    • Mangray and King, Front Biosci., 3:D1148-1160, 1998.
    • Masuda C, et al. Jpn J Cancer Res. 2000, 91(6):582-8.
    • Mayer, Radiat Oncol Investig. 6:281-8, 1998.
    • Mumby and Walter, Cell Regul., 2:589-598, 1991.
    • Nagata et al., J. Immunol. 1997, 159:1336
    • Nanni et al., J. Exp. Med. 2001, 194:1195.
    • Natoli et al., Biochem. Pharmacol., 56(8):915-920, 1998.
    • Ohara, Acta Oncol. 37:471-4, 1998.
    • Pardoll D, Nature Reviews Immunol., 2002, 2:227-238.
    • Peoples et al., Proc. Natl. Aca. Sci., 92:6547-511995, 1995.
    • Rovero et al., J. immuno., 2000, 165:5133-5142
    • Sabel M S, et al., Am. J. Clin. Dermatol. 2002, 3:609-16
    • Shau H, et al., J Leukocyte Biol. 1992; 51:343-9.
    • Solyanik et al., Cell Prolif, 28:263-278, 1995.
    • Spik G, et al., Adv Exp Med Biol. 1994; 357:13-9.
    • Stokke et al., Cell Prolif, 30(5):197-218, 1997.
    • Tanaka T, et al. Jpn J Cancer Res. 2000, 91(1):25-33.
    • Tsuda H, et al., Biofactors. 2000; 12(1-4):83-8.
    • Ushida Y, et al. Jpn J Cancer Res. 1999, 90(3):262-7.
    • Vaiashampayan et.al., Clin. Cancer Res. 2002, 8:3696-701
    • Wang W P, et al., Jpn J Cancer Res. 2000, 91(10):1022-7.
    • Yoo Y C, et al., Adv Exp Med Biol. 1998, 443:285-91.
    • Yoo Y C, et al., Jpn J Cancer Res. 1997, 88(2):184-90.
  • 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 description. 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 descriptions are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or steps.

Claims (16)

1. A method of treating cancer comprising the step of administering to a subject a cancer immunotherapy and an adjuvant, wherein said adjuvant is a lactoferrin composition that is administered orally for gastrointestinal delivery in an amount sufficient to provide an improvement in the cancer in said subject and wherein any N-terminal lactoferrin variant in the lactoferrin composition has a deletion, substitution, or combination thereof, from 1 to 16 N-terminal amino acid residues and the N-terminal lactoferrin variant retains the same biological function as full length lactoferrin, wherein said lactoferrin composition comprises recombinant bovine lactoferrin or recombinant human lactoferrin.
2. The method of claim 1, wherein said lactoferrin composition is dispersed in a pharmaceutically acceptable carrier.
3. The method of claim 1, wherein any N-terminal lactoferrin variant in the lactoferrin composition lacks at least the N-terminal glycine residue.
4. The method of claim 1, wherein the lactoferrin composition comprises an amount of the lactoferrin and/or N-terminal lactoferrin variant such that about 10 mg to about 100 g of the lactoferrin and/or N-terminal lactoferrin variant is administered per day.
5. The method of claim 1, wherein the immunotherapy comprises administration of a nucleic acid sequence expressing a cancer antigen to the subject, wherein expression of the antigen is transient.
6. The method of claim 5, wherein the nucleic acid sequence is contained in a vector, wherein expression of the antigen is transient.
7. The method of claim 1, wherein the immunotherapy comprises administration of a vector containing a nucleic acid sequence expressing an immunomodulatory cytokine to the subject, wherein expression of the immunomodulatory cytokine is transient.
8. A method of treating cancer comprising the step of administering to a subject a cancer immunotherapy and an adjuvant, wherein said adjuvant is a lactoferrin composition that is administered orally for gastrointestinal delivery in an amount sufficient to provide an improvement in the cancer in said subject and wherein any N-terminal lactoferrin variant in the lactoferrin composition has a deletion, substitution, or combination thereof, from 1 to 16 N-terminal amino acid residues and the N-terminal lactoferrin variant retains the same biological function as full length lactoferrin, wherein the immunotherapy comprises administering antigen presenting cells.
9. The method of claim 8, wherein the cells are allogeneic.
10. The method of claim 8, wherein the cells are syngeneic.
11. A method of treating cancer comprising the step of administering to a subject a cancer immunotherapy and an adjuvant, wherein said adjuvant is a lactoferrin composition that is administered orally for gastrointestinal delivery in an amount sufficient to provide an improvement in the cancer in said subject and wherein any N-terminal lactoferrin variant in the lactoferrin composition has a deletion, substitution, or combination thereof, of from 1 to 16 N-terminal amino acid residues and the N-terminal lactoferrin variant retains the same biological function as full length lactoferrin, wherein the lactoferrin composition is administered sequentially with the immunotherapy.
12. A method of enhancing the immune system in a subject suffering from cancer comprising the step of administering to the subject a cancer immunotherapy and an adjuvant, wherein the adjuvant is an orally administered lactoferrin composition and wherein any N-terminal lactoferrin variant in the lactoferrin composition has a deletion, substitution, or combination thereof, of from 1 to 16 N-terminal amino acid residues and the N-terminal lactoferrin variant retains the same biological function as full length lactoferrin, wherein said lactoferrin composition comprises recombinant human lactoferrin or recombinant bovine lactoferrin.
13. The method of claim 12, wherein said lactoferrin composition_stimulates the production of interleukin-18, GM-CSF, or MIP-3alpha.
14. The method of claim 13, wherein said interleukin-18, GM-CSF or MIP-3alpha stimulate the production, maturation, migration or activity of immune cells.
15. The method of claim 12, wherein the immune cells are T lymphocytes, natural killer cells, dendritic cells, antigen presenting cells or progenitor cell.
16. The method of claim 15, wherein the T lymphocytes are selected from the group consisting of CD4+, CD8+ and CD3+ cells.
US12/555,296 2003-06-06 2009-09-08 Lactoferrin as an adjuvant in cancer vaccines Abandoned US20110110983A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/555,296 US20110110983A1 (en) 2003-06-06 2009-09-08 Lactoferrin as an adjuvant in cancer vaccines

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US47631803P 2003-06-06 2003-06-06
US49823603P 2003-08-27 2003-08-27
US10/862,213 US8105615B2 (en) 2003-06-06 2004-06-07 Lactoferrin as an adjuvant in cancer vaccines
US12/555,296 US20110110983A1 (en) 2003-06-06 2009-09-08 Lactoferrin as an adjuvant in cancer vaccines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/862,213 Continuation US8105615B2 (en) 2003-06-06 2004-06-07 Lactoferrin as an adjuvant in cancer vaccines

Publications (1)

Publication Number Publication Date
US20110110983A1 true US20110110983A1 (en) 2011-05-12

Family

ID=34594508

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/862,213 Expired - Fee Related US8105615B2 (en) 2003-06-06 2004-06-07 Lactoferrin as an adjuvant in cancer vaccines
US12/555,296 Abandoned US20110110983A1 (en) 2003-06-06 2009-09-08 Lactoferrin as an adjuvant in cancer vaccines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/862,213 Expired - Fee Related US8105615B2 (en) 2003-06-06 2004-06-07 Lactoferrin as an adjuvant in cancer vaccines

Country Status (6)

Country Link
US (2) US8105615B2 (en)
EP (1) EP1635766A4 (en)
JP (2) JP2007524654A (en)
AU (1) AU2004289170B2 (en)
CA (1) CA2527904A1 (en)
WO (1) WO2005046571A2 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003099323A1 (en) * 2002-05-10 2003-12-04 Agennix Incorporated Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
CA2499014A1 (en) 2002-09-16 2004-03-25 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
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
US7323443B2 (en) * 2002-12-12 2008-01-29 Agennix Inc. Lactoferrin in the reduction of pain
JP2007524654A (en) * 2003-06-06 2007-08-30 エイジェニックス インコーポレイテッド Lactoferrin as an adjuvant in cancer vaccines
WO2006047744A2 (en) * 2004-10-26 2006-05-04 Agennix Incorporated Compositions of lactoferrin related peptides and uses thereof
EP1835930A4 (en) * 2004-11-19 2012-11-21 Fonterra Corporate Res And Dev Ltd Methods of immune or haematological enhancement, inhibiting tumour formation or growth, and treating or preventing cancer
MX2009012858A (en) 2007-06-01 2010-02-03 Jackson H M Found Military Med Vaccine for the prevention of breast cancer relapse.
US20130344003A1 (en) * 2012-06-25 2013-12-26 The Board Of Trustees Of The Leland Stanford Junior University Immuno-therapy for cancer treatment using iron oxide nanoparticles

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4791193A (en) * 1986-07-17 1988-12-13 Morinaga Milk Industry Co., Ltd. Process for producing bovine lactoferrin in high purity
US4977137A (en) * 1987-06-03 1990-12-11 Baylor College Of Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US5006333A (en) * 1987-08-03 1991-04-09 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
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
US5466669A (en) * 1992-03-07 1995-11-14 Morinaga Milk Industry Co., Ltd. Immunostimulatory agent
US5571896A (en) * 1992-04-24 1996-11-05 Baylor College Of Medicine Production of recombinant human lactoferrin
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
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
US5712247A (en) * 1995-02-21 1998-01-27 University Of North Carolina Use of lactoferrin to modulate and/or neutralize heparin activity
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
US5833975A (en) * 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
US5849881A (en) * 1989-05-05 1998-12-15 Baylor College Medicine Production of recombinant lactoferrin and lactoferrin polypeptides using cDNA sequences in various organisms
US5919913A (en) * 1994-02-16 1999-07-06 Pharming B.V. Isolation of lactoferrin from milk
US6066469A (en) * 1990-03-08 2000-05-23 Ferro Dynamics, Inc. Cloning, expression, and uses of human lactoferrin
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
US20010007659A1 (en) * 1997-04-17 2001-07-12 The University Of California Use of lentiviral vectors for antigen presentation in dendritic cells
US20010036928A1 (en) * 1996-04-22 2001-11-01 Chamberlain Ronald S. Heterologous boosting immunizations
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
US20020072596A1 (en) * 1999-12-23 2002-06-13 Ruben Steven M. Transferrin polynucleotides, polypeptides, and antibodies
US20020198362A1 (en) * 2000-03-01 2002-12-26 Alexander Gaiger Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
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
US20030203839A1 (en) * 1999-10-29 2003-10-30 Kruzel Marian L. Immune enhancing composition containing lactoferrin
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
US20040023334A1 (en) * 2001-08-30 2004-02-05 Biorexis Pharmaceutical Corporation Modified transferrin fusion proteins
US20040142037A1 (en) * 2002-09-16 2004-07-22 Jose Engelmayer Lactoferrin compositions and methods of wound treatment
US20040151784A1 (en) * 2002-12-12 2004-08-05 Atul Varadhachary Lactoferrin in the reduction of pain
US20040152623A1 (en) * 2002-12-04 2004-08-05 Atul Varadhachary Lactoferrin in the reduction of circulating cholesterol, vascular inflammation, atherosclerosis and cardiovascular disease
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
US20050004006A1 (en) * 2003-05-14 2005-01-06 Jose Engelmayer Lactoferrin in the treatment of diabetes mellitus
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

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6351337A (en) 1986-08-22 1988-03-04 Snow Brand Milk Prod Co Ltd Antitumor agent
JP3184923B2 (en) 1992-01-08 2001-07-09 ビオ セレ ラボラトワール エス ア Anti-rheumatic drug
JP3207647B2 (en) * 1993-12-21 2001-09-10 森永乳業株式会社 Immunostimulants that promote production of IgG and IgM class antibodies
JPH08217693A (en) 1995-02-17 1996-08-27 Yoshihisa Naito New medicine composition
US20020016289A1 (en) * 1995-06-01 2002-02-07 Orla M. Conneely Methods for treatment and prevention of helicobacter pylori infection using lactoferrin
JP3496387B2 (en) 1996-01-23 2004-02-09 花王株式会社 Hair cosmetics
JP2001504447A (en) 1996-08-12 2001-04-03 エイプラス サイエンス インベスト アーベー Treatment and prevention of infection, inflammation and / or tumor with lactoferrin and / or lactoferricin
JPH1059864A (en) 1996-08-15 1998-03-03 Morinaga Milk Ind Co Ltd Cancer metastasis suppressing agent for oral administration
WO1999052545A1 (en) * 1998-04-13 1999-10-21 New England Medical Center Hospitals, Inc. Methods and compositions for inactivating infectious agents using lactoferrin and related molecules
JP2002519332A (en) 1998-06-26 2002-07-02 エヌ・ヴェー・ニュートリシア Pharmaceutical preparation for use in treating or preventing surface infections caused by microorganisms
US6660275B2 (en) * 1998-07-27 2003-12-09 Aventis Pasteur Limited Chlamydia antigens and corresponding DNA fragments and uses thereof
IL144371A0 (en) * 1999-01-29 2002-05-23 Corixa Corp Her-2/neu fusion proteins
JP2000229881A (en) 1999-02-10 2000-08-22 Morinaga Milk Ind Co Ltd Prognosis-improving agent for cancerous disease
AUPQ347199A0 (en) * 1999-10-15 1999-11-11 Csl Limited Novel polypeptide fragments
EP1385541B1 (en) * 2000-04-13 2008-06-18 Corixa Corporation Immunostimulant compositions comprising an aminoalkyl glucosaminide phosphate and qs-21
RU2165769C1 (en) 2000-07-13 2001-04-27 Якубовская Раиса Ивановна Antibacterial, antioxidant, immunomodulating and anticancer preparation and method of its embodiment
EP1835930A4 (en) 2004-11-19 2012-11-21 Fonterra Corporate Res And Dev Ltd Methods of immune or haematological enhancement, inhibiting tumour formation or growth, and treating or preventing cancer
JP2007233064A (en) 2006-03-01 2007-09-13 Bitsign:Kk Display system and display device
NZ552316A (en) 2006-12-22 2009-10-30 Fonterra Co Operative Group Dairy product and process

Patent Citations (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4791193A (en) * 1986-07-17 1988-12-13 Morinaga Milk Industry Co., Ltd. Process for producing bovine lactoferrin in high purity
US4977137A (en) * 1987-06-03 1990-12-11 Baylor College Of Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US4977137B1 (en) * 1987-06-03 1994-06-28 Baylor College Medicine Lactoferrin as a dietary ingredient promoting the growth of the gastrointestinal tract
US5006333A (en) * 1987-08-03 1991-04-09 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
US5833975A (en) * 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
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
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
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
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
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
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
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
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
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
US6066469A (en) * 1990-03-08 2000-05-23 Ferro Dynamics, Inc. Cloning, expression, and uses of human lactoferrin
US6277817B1 (en) * 1990-03-08 2001-08-21 Ferro Dynamics Inc. Human lactoferrin
US6455687B1 (en) * 1990-03-08 2002-09-24 Ferradynamics, Inc Human lactoferrin
US5466669A (en) * 1992-03-07 1995-11-14 Morinaga Milk Industry Co., Ltd. Immunostimulatory agent
US5571896A (en) * 1992-04-24 1996-11-05 Baylor College Of Medicine Production of recombinant human lactoferrin
US5919913A (en) * 1994-02-16 1999-07-06 Pharming B.V. Isolation of lactoferrin from milk
US5712247A (en) * 1995-02-21 1998-01-27 University Of North Carolina Use of lactoferrin to modulate and/or neutralize heparin activity
US20010036928A1 (en) * 1996-04-22 2001-11-01 Chamberlain Ronald S. Heterologous boosting immunizations
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
US20010007659A1 (en) * 1997-04-17 2001-07-12 The University Of California Use of lentiviral vectors for antigen presentation in dendritic cells
US6890902B2 (en) * 1998-08-28 2005-05-10 Alpharma As Cytotoxic modified lactoferrin peptides
US20030022821A1 (en) * 1998-08-28 2003-01-30 Alpharma As Bioactive peptides
US6399570B1 (en) * 1999-02-05 2002-06-04 Agennix, Inc. Antimicrobial/endotoxin neutralizing polypeptide
US20030105006A1 (en) * 1999-02-05 2003-06-05 Mann David M. Antimicrobial/endotoxin neutralizing polypeptide
US20030203839A1 (en) * 1999-10-29 2003-10-30 Kruzel Marian L. Immune enhancing composition containing lactoferrin
US20020072596A1 (en) * 1999-12-23 2002-06-13 Ruben Steven M. Transferrin polynucleotides, polypeptides, and antibodies
US20020198362A1 (en) * 2000-03-01 2002-12-26 Alexander Gaiger Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20030096736A1 (en) * 2001-05-09 2003-05-22 Kruzel Marian L. Lactoferrin for age related disorders in humans
US20040023334A1 (en) * 2001-08-30 2004-02-05 Biorexis Pharmaceutical Corporation Modified transferrin fusion proteins
US20040009895A1 (en) * 2002-05-10 2004-01-15 Atul Varadhachary Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20040082504A1 (en) * 2002-05-10 2004-04-29 Atul Varadhachary Intratumorally administered lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
US20100137208A1 (en) * 2002-05-10 2010-06-03 Atul Varadhachary Intratumorally administered lactoferrin in the treatment of malignantneoplasms 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
US20040152623A1 (en) * 2002-12-04 2004-08-05 Atul Varadhachary Lactoferrin in the reduction of circulating cholesterol, vascular inflammation, atherosclerosis and cardiovascular disease
US7026295B2 (en) * 2002-12-04 2006-04-11 Agennix Incorporated Lactoferrin in the reduction of circulating cholesterol, vascular inflammation, atherosclerosis and cardiovascular disease
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
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
US7323443B2 (en) * 2002-12-12 2008-01-29 Agennix Inc. Lactoferrin in the reduction of pain
US20040151784A1 (en) * 2002-12-12 2004-08-05 Atul Varadhachary Lactoferrin in the reduction of pain
US20050004006A1 (en) * 2003-05-14 2005-01-06 Jose Engelmayer Lactoferrin in the treatment of diabetes mellitus
US7034126B2 (en) * 2003-05-14 2006-04-25 Agennix, Inc. Lactoferrin in the treatment of diabetes mellitus
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

Also Published As

Publication number Publication date
US20050019342A1 (en) 2005-01-27
EP1635766A2 (en) 2006-03-22
AU2004289170B2 (en) 2009-12-17
JP2007524654A (en) 2007-08-30
US8105615B2 (en) 2012-01-31
WO2005046571A3 (en) 2008-01-17
AU2004289170A1 (en) 2005-05-26
WO2005046571A2 (en) 2005-05-26
CA2527904A1 (en) 2005-05-26
JP2012097109A (en) 2012-05-24
EP1635766A4 (en) 2009-09-30

Similar Documents

Publication Publication Date Title
US20110110983A1 (en) Lactoferrin as an adjuvant in cancer vaccines
JP5156084B2 (en) Lactoferrin in the treatment of established solid tumors
Maverakis et al. Metastatic melanoma–a review of current and future treatment options
KR100704127B1 (en) Active Antiangiogenic Therapy
PT1659178E (en) A process for the purification or production of a mage protein
Ben-Efraim One hundred years of cancer immunotherapy: a critical appraisal
CA2477780A1 (en) Methods of enhancing immune induction involving mda-7
PT2035581E (en) Dna composition against tumor stromal antigen fap and methods of use thereof
Huang et al. A cancer vaccine based on the marine antimicrobial peptide pardaxin (GE33) for control of bladder-associated tumors
Shi et al. Granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted by cDNA-transfected tumor cells induces a more potent antitumor response than exogenous GM-CSF
US20140205609A1 (en) Methods for inducing systemic immune responses to cancer
CN104853764A (en) MSI-specific frameshift peptides (FSP) for prevention and treatment of cancer
di Pietro et al. Oncophage®(vitespen®)-Heat shock protein peptide complex 96-based vaccines in melanoma: How far we are, how far we can get?
WO2013048226A1 (en) Immunomodulator metallopeptides (immps) and compositions containing same
JP2005247861A (en) Mucin peptide having immuno-enhancing characteristic
JP2003306444A (en) Immunoadjuvant
Maverakis et al. Please let us know how this document benefits you.
BRPI0400755A2 (en) use of polypeptides and / or oligonucleotides, and vaccine composition

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGENNIX INCORPORATED, TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PERICLE, FEDERICA;VARADHACHARY, ATUL;REEL/FRAME:027468/0876

Effective date: 20040923

STCB Information on status: application discontinuation

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