EP1556074A2 - Peptides derives de la caseine et leurs utilisations therapeutiques - Google Patents

Peptides derives de la caseine et leurs utilisations therapeutiques

Info

Publication number
EP1556074A2
EP1556074A2 EP02758768A EP02758768A EP1556074A2 EP 1556074 A2 EP1556074 A2 EP 1556074A2 EP 02758768 A EP02758768 A EP 02758768A EP 02758768 A EP02758768 A EP 02758768A EP 1556074 A2 EP1556074 A2 EP 1556074A2
Authority
EP
European Patent Office
Prior art keywords
peptide
pharmaceutical composition
derived
casein
terminus portion
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.)
Withdrawn
Application number
EP02758768A
Other languages
German (de)
English (en)
Other versions
EP1556074A4 (fr
Inventor
Zvi Sidelman
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.)
Chay 13 Medical Research Group Nv
Chay 13 Medical Res Group NV
Original Assignee
Chay 13 Medical Research Group Nv
Chay 13 Medical Res Group NV
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
Priority claimed from US09/942,121 external-priority patent/US20020147144A1/en
Application filed by Chay 13 Medical Research Group Nv, Chay 13 Medical Res Group NV filed Critical Chay 13 Medical Research Group Nv
Publication of EP1556074A2 publication Critical patent/EP1556074A2/fr
Publication of EP1556074A4 publication Critical patent/EP1556074A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4732Casein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • the present invention relates to biologically active peptides that are derived from or are similar to sequences identical with the N-terminus of the ⁇ SI fraction of milk casein. These peptides are capable of stimulating and enhancing immune response, protecting against viral infection, normalizing serum cholesterol levels, and stimulating hematopoiesis.
  • the casein-derived peptides are non-toxic and can be used to treat and prevent immune pathologies, hypercholesterolemia, hematological disorders and viral-related diseases.
  • Bioactive molecules from nutrients In addition to the nutritional value of many foods, certain fractions and products of digestive pathways possess the ability to influence physiological processes. Some of these "extranutritional" constituents are present in their active form in the whole nutriment, such as the immunoglobulins in mother's milk and colostrums, phytoestrogens found in soy-based foods, polyphenolic antioxidants from fruits and vitamins. Others are encrypted within nutrient molecules, and are released in an active form during digestion or food processing, for example antihypertensive peptides from lactoglobin [Kitts, D. D. (1999), Can. J. Physiol. Pharmacol. 72:4; 423-434].
  • Casein the predominant milk protein, has been traditionally defined as composed of three fractions, ⁇ , ⁇ and ⁇ , according to their electrophoretic mobility [N. J. Hipp, et al. (1952), Dairy Sci., 35:272].
  • Today casein is defined according to the amino acid sequences of each of the subgroups ⁇ Sl , ⁇ S2, ⁇ and ⁇ [W. N. Engel et ⁇ /. (1984), J. Dairy Sci. 67: 1599].
  • casein proteins are subjected to proteolytic cleavage by acid proteases such as chymosin (rennin), trypsin and pepsin, producing shorter peptides and causing curdling and calcium sequestration by the resultant protein fragments.
  • acid proteases such as chymosin (rennin), trypsin and pepsin
  • U.S. Patent No. 3,764,670 discloses proteolytic casein digests possessing antibiotic properties against microorganisms.
  • Israel Patent No. 42863 describes a casein-derived peptide consisting of 23 amino acids of the N-terminus of casein, possessing anti-bacterial activity.
  • other physiologically active properties such as opioid and growth factor-like activities have been proposed for casein or its derivatives [Kitts, D. D., (1999), ibid.).
  • G-CSF granulocyte colony stimulation factor
  • GM-CSF granulocyte macrophage colony stimulating factor
  • thrombocytopenia Following high dose chemotherapy and especially following ASCT, patients are at risk for thrombocytopenia which may last for many months even up to 3 years and some thromboctyopenic patients may never recover. Many patients previously treated with multiple blood products become platelet resistant and hence thrombocytopenia may be impossible to overcome, even transiently, despite intensive and frequent platelet transfusions from a single donor. Resistance to platelets and protracted thrombocytopenia represent a common cause of death at ASCT centers worldwide.
  • rhIL3 recombinant human interleukin-3
  • rhIL6 recombinant human interleukin-6
  • thrombopoetin (TPO) in regulation of hematopoiesis and platelet function:
  • TPO appears to be the major regulator of platelet production in vivo, although increase in the kidney- and liver-derived growth factor in platelet deficiencies is not caused by adaptation of TPO biosynthesis in these organs. Rather, a "feed-back loop" seems to exist in which the number of circulating platelets determines how much of the circulating TPO is available to the bone marrow for platelet production.
  • TPO is an early acting cytokine with important multilineage effects: TPO alone, or in combination with other early acting cytokines, can (i) promote viability and suppress apoptosis in progenitor cells; (ii) regulate hematopoietic stem cell production and function; (iii) trigger cell division of dormant multipotent cells; (iv) induce multilineage differentiation and (v) enhance formation of multilineage colonies containing granulocytes, erythrocytes, macrophages, and megakaryocytes (MK, CFU-GEMM).
  • MK megakaryocytes
  • TPO stimulates the production of more limited progenitors for granulocyte/monocyte, megakaryocyte and erythroid colonies, and stimulates adhesion of primitive human bone marrow and megakaryocytic cells to fibronectin and fibrinogen.
  • TPO is an important cytokine for clinical hematologists/transplanters: for the mobilization, amplification and ex vivo expansion of stem cells and committed precursor cells for autologous and allogeneic transplantation [von dem Borne, A.E.G.Kr., et al., (1998) Thrombopoietin: it's role in platelet disorders and as a new drug in clinical medicine. In Ba Amsterdam Clin. Hematol. June: l 1(2), 427-45].
  • this potent growth factor primes platelets for various agonists and modulates platelet-extracellular matrix interactions. Although it does not itself cause platelet aggregation, TPO upregulates ADP-induced aggregation, especially on the second wave of aggregation, upregulates granule (ADP, ATP, serotonin, etc.) release and production of thromboxane B2, increases platelet attachment to collagen and potentiates shear-induced platelet aggregation. TPO also stimulates PMN activation, inducing IL-8 release and priming oxygen metabolite production, likely enhancing antimicrobial defense.
  • TPO idiopathic aplastic anemia
  • AA idiopathic aplastic anemia
  • TPO is elevated in other forms of aplastic thrombocytopenia as well, but not in conditions of increased platelet destruction.
  • the reactive increase in TPO production is insufficient in cases of destructive thrombocytopenia.
  • TPO is not only a therapeutic option for aplastic, but also for destructive thrombocytopenia.
  • Thrombopoietic agents are of great clinical interest, for prevention and/or treatment of pathological or treatment-induced thrombocytopenia, and as a substitute for platelet transfusions.
  • cytokines Of the cytokines evaluated, all but the marginally potent IL-11 have been deemed unacceptable for clinical use.
  • TPO is widely believed to become the cytokine of choice for throbocytopenia treatment.
  • Recombinant human TPO (Genentech) has recently become available, enabling accurate pharmacokinetic determinations and clinical trials.
  • TPO's potential applications encompass the realms of supportive care (post chemo/radio-therapy, bone marrow and stem cell transplantation), hematological disease (AA, myelodysplasia, congenital and acquired thrombocytopenia), liver diseases, transfusion (expansion, harvest, mobilization and storage of platelets) and surgery (including liver transplantation).
  • supportive care post chemo/radio-therapy, bone marrow and stem cell transplantation
  • AA myelodysplasia
  • congenital and acquired thrombocytopenia liver diseases
  • transfusion expansion, harvest, mobilization and storage of platelets
  • surgery including liver transplantation.
  • TPO/EPO/G-CSF cocktail for myelodysplasia, G-CSF and TPO combination for peripheral stem cell mobilization and TPO in harvesting CD 34+ cells and ex vivo expansion of megakaryocytes for superior platelet reconstitution.
  • TPO is costly and potentially antigenic at therapeutically effective levels.
  • the ⁇ Sl fraction of casein can be obtained from milk proteins by various methods [D. G. Schmidth and T. A. J. Paynes (1963), Biochim., Biophys. Acta, 78:492; M. P. Thompson and C. A. Kiddy (1964), J. Dairy Sci., 47:626; J. C. Mercier, et al. (1968), Bull. Soc. Chi . Biol. 50:521], and the complete amino acid sequence of the ⁇ S 1 fraction of casein was determined by J. C. Mercier et al. (1971) (Eur. J. Biochem. 23:41).
  • Dairy Res., 60:401] as has the intestinal absorption and appearance of this fragment in mammalian plasma following ingestion of whole milk proteins [Fiat, A.M., et al. (1998) Biochimie, 80(2):2155-65].
  • Meisel, H. and Bockelmann, W. [(1999), Antonie Van Leeuwenhoek, 76:207-15] detected amino acid sequences of immunopeptides, casokinins and casomorphins in peptides liberated by lactic acid bacteria digests of ⁇ and ⁇ casein fractions.
  • Of particular interest is the anti-aggregating and thrombolytic activity demonstrated for C-terminal portions of the ⁇ - and ⁇ -casein fractions [Chabance, B. et al. (1997), Biochem. Mol. Biol. Int. 42(1) 77-84; Fiat AM. et al. (1993), J. Dairy Sci. 76(1): 301-310].
  • a method of preventing or treating an autoimmune disease the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating a viral disease the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing viral infection the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of inducing hematopoiesis the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing hematopoietic stem cells proliferation the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing hematopoietic stem cells proliferation and differentiation the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing megakaryocytopoiesis the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing erythropoiesis the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S I casein.
  • a method of inducing leukocytopoiesis the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing thrombocytopoiesis the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing plasma cell proliferation the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing dendritic cell proliferation the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing macrophage proliferation the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating thrombocytopenia the method effected by administering 5 to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating pancytopenia the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived l o from an N terminus portion of ⁇ S 1 casein.
  • a method of preventing or treating granulocytopenia the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating hyperlipidemia the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of 0 preventing or treating cholesteremia the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating glucosuria the method effected by administering to a 5 subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating diabetes the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of preventing or treating AIDS the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating infection by HIV the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a method of treating an erythropoietin treatable condition the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of augmenting the effect of erythropoietin the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of treating a thrombopoietin treatable condition the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of augmenting the effect of thrombopoietin the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of enhancing peripheral stem cell mobilization the method effected by administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising effective amounts of thrombopoietin and a peptide derived from an N terminus portion of ⁇ SI casein.
  • a pharmaceutical composition for preventing or treating an autoimmune disease the pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating a viral disease comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing viral infection comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ S 1 casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation and differentiation comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing megakaryocytopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing erythropoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing Ieukocytopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing thrombocytopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing plasma cell proliferation comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing dendritic cell proliferation comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing macrophage proliferation comprising a peptide derived from an N terminus portion of ⁇ S 1 casein and a pharmaceutically acceptable carrier.
  • a pha ⁇ naceutical composition for preventing or treating thrombocytopenia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating pancytopenia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating granulocytopenia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating hyperlipidemia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating cholesteremia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating glucosuria comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating diabetes comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating AIDS comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating infection by HIV comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a pharmaceutical composition for treating a thrombopoietin treatable condition comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for augmenting the effect of thrombopoietin comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for enhancing peripheral stem cell mobilization comprising, as active ingredients thrombopoietin and a peptide derived from an N terminus portion of ⁇ SI casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation and differentiation comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ccS 1 casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing megakaryocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing erythropoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing Ieukocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing thrombocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating thrombocytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating pancytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating granulocytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for treating or preventing an indication selected from the group consisting of hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, myelodysplastic syndrome, aplastic anemia and bone marrow insufficiency, the pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating a donor of said donated blood stem cells with a peptide derived from an N terminus portion of ⁇ Sl casein prior to donation and implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating said donated blood stem cells with a peptide derived from an N terminus portion of ⁇ Sl casein prior to implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of blood stem cells in a myeloablated recipient the method effected by treating said blood stem cells with a peptide derived from an N terminus portion of ⁇ Sl casein prior to implanting the blood stem cells in the recipient.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating a donor of said donated blood stem cells with a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin prior to donation and implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating said donated blood stem cells with a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin prior to implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of blood stem cells in a myeloablated recipient the method effected by treating said blood stem cells with a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin prior to implanting the blood stem cells in the recipient.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating an autoimmune disease.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating a viral disease.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing viral infection.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for inducing hematopoiesis.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing hematopoietic stem cells proliferation.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing hematopoietic stem cells proliferation and differentiation. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing megakaryocytopoiesis.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing erythropoiesis.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing Ieukocytopoiesis. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing thrombocytopoiesis.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing dendritic cell proliferation.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for inducing macrophage proliferation.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating thrombocytopenia. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating pancytopenia. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating granulocytopenia.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating hyperlipidemia.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating cholesteremia.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating glucosuria.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating diabetes.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating AIDS.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for the preparation of a medicament for preventing or treating infection by HIV.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for preventing or treating conditions associated with myeloablative doses of chemoradiotherapy supported by autologous bone marrow or peripheral blood stem cell transplantation (ASCT) or allogeneic bone marrow transplantation (BMT).
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for treating a thrombopoietin treatable condition.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for enhancing peripheral stem cell mobilization. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for the preparation of a medicament for enhancing colonization of blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an autoimmune disease.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating a viral disease.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating a viral infection.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cell proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation and differentiation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing megakaryocytopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing erythropoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing Ieukocytopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing thrombocytopoiesis. Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing plasma cell proliferation. Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing dendritic cell proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing macrophage proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating thrombocytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating pancytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating granulocytopenia. Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating hyperlipidemia. Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating cholesteremia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating glucosuria.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating diabetes.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating AIDS.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating infection by HIV.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating conditions associated with myeloablative doses of chemoradiotherapy supported by autologous bone marrow or peripheral blood stem cell transplantation (ASCT) or allogeneic bone marrow transplantation (BMT).
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for treating a thrombopoietin treatable condition.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for augmenting the effect of thrombopoietin.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier peptide derived from an N terminus portion of ⁇ Sl casein for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier enhancing colonization of blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for enhancing peripheral stem cell mobilization.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation and differentiation.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing megakaryocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing erythropoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing Ieukocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing thrombocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating thrombocytopenia.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating pancytopenia.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating granulocytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an indication selected from the group consisting of autoimmune disease or condition, viral disease, viral infection, hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, hyperlipidemia, hypercholesterolemia, glucosuria, hyperglycemia, diabetes, AIDS, HIV-1, helper T-cell disorders, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, immune system disorders resulting from chemotherapy or radiation therapy, human immune system disorders resulting from treatment of diseases of immune deficiency and bacterial infections.
  • an indication selected from the group consisting of autoimmune disease or condition, viral disease, viral infection, hematological disease, hematological deficiencies, thro
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an indication selected from the group consisting of hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, myelodysplastic syndrome, aplastic anemia and bone marrow insufficiency.
  • a pharmaceutical composition comprising, as active ingredients, a peptide derived from an N terminus portion of ⁇ Sl casein and thrombopoietin, and a pharmaceutically acceptable carrier for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as active ingredients, a peptide derived from an N terminus portion of ⁇ Sl casein and thrombopoietin, and a pharmaceutically acceptable carrier for enhancing colonization of blood stem cells in a myeloablated recipient.
  • the peptide is a fragment derived by fragmentation of ⁇ Sl casein.
  • the peptide is a synthetic peptide.
  • the peptide has a sequence as set forth in one of SEQ ID NOs:l-25.
  • a purified peptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-25 Further according to the present invention there is provided a pharmaceutical composition comprising a purified peptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-25 and a pharmaceutically acceptable carrier. Further according to the present invention there is provided a pharmaceutical composition comprising thrombopoietin and a purified peptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-25 and a pharmaceutically acceptable carrier.
  • the present invention successfully addresses the shortcomings of the presently known configurations by providing peptides for the treatment of human disease, which peptides are derived from the N terminus portion of ⁇ Sl casein and posses no detectable toxicity and high therapeutic efficacy.
  • FIG. 1 depicts the stimulation of Natural Killer (NK) cell activity in cultured murine bone marrow cells by peptides derived from natural casein. Lysis of S labeled YAC target cells by cultured murine bone marrow cells incubated in the presence or absence of 100 ⁇ g per ml peptides derived from natural casein is expressed as the fraction of total radioactivity released from the YAC cells into the culture supernatant ( % Release S).
  • Figure 1 represents NK activity at an effecto ⁇ target cell ratio of 25:1 and 50: 1.
  • FIGs. 2a and 2b depict the stimulation of Natural Killer (NK) cell activity in cultured human Peripheral Blood Stem Cells (PBSC) by peptides derived from natural casein.
  • PBSC Peripheral Blood Stem Cells
  • FIG. 2a represents NK activity of two blood samples from the same patient, incubated at different effecto ⁇ target cell ratios (1 :25 and 1 :50).
  • Figure 2b represents NK activity of blood samples from normal and affected donors incubated at the same effecto ⁇ target cell ratio. Squares represent an effecto ⁇ target cell ratio of 100: 1, diamonds represent an effector :target cell ratio of 50: 1.
  • FIGs. 3a-3c depict the stimulation of proliferation of Natural Killer (NK) and T-lymphocyte (T) cells from cultured human Peripheral Blood Stem Cells (PBSC) by peptides derived from natural casein.
  • NK and T cell proliferation in cultured PBSC from Granulocyte Colony Stimulating Factor treated donors incubated with or without peptides derived from natural casein is expressed as the percentage (%) of cells binding the anti-CD 3 /FITC fluorescent anti-T cell antibody UCHT,, or the anti CD 56 RPE fluorescent anti-NK cell antibody MOC-1 (DAKO A/S Denmark).
  • Controls are FITC and RPE-conjugated anti-mouse IgG antibody.
  • Figure 3a represents the percentage of cultured human PBSC binding fluorescent antibody CD 56 (5 independent samples) after 10 days incubation with (peptides) or without (control) 100 ⁇ g per ml peptides derived from natural casein.
  • Figure 3b represents the percentage of cultured human PBSC cells binding fluorescent anti-CD 3 (T cell) antibody, following 14 days of incubation with (peptides) or without (control) 100 ⁇ g per ml peptides derived from natural casein.
  • Figure 3c represents the percentage of cultured human PBSC cells binding fluorescent anti-CD 3 (T cell) antibody and cells binding both CD 3 and CD 56 (T and NK-like cells) antibodies after 28 days incubation with (peptides) or without (control) 100 ⁇ g per ml peptides derived from natural casein.
  • FIG. 4 depicts the stimulation of Natural Killer (NK) cell activity in cultured human Peripheral Blood Stem Cells (PBSC) by synthetic peptides derived from casein. Lysis of S labeled K562 target cells by cultured human PBSC (from a breast cancer patient) incubated without (0 ⁇ g) or with increasing concentrations (10 - 500 ⁇ g per ml) of synthetic peptides derived from casein is expressed as the fraction of total radioactivity released from the K562 cells into the culture supernatant (% Release).
  • NK Natural Killer
  • Peptides represent N-terminal sequences of 1-10 (la, diamonds), 1-11 (2a, squares) and 1-12 (3a, triangles) first amino acids of the N terminus portion of ⁇ Sl casein (see Table 3 below for sequences of synthetic peptides).
  • FIGs. 5a-5c depict the stimulation of proliferation of cultured human cells of diverse origin by peptides derived from natural casein. Proliferation of the cultured human cells after 14-21 days incubation with increasing concentrations of the peptides derived from natural casein is expressed as the amount of [ H]-thymidine incorporated into each sample.
  • Figure 5a represents the incorporation of label into two samples (PBSC 1, squares, 15 days incubation; and PBSC 2, diamonds, 20 days incubation) of human Peripheral Blood Stem Cells incubated with or without (ctrl) 50 - 600 ⁇ g per ml peptides derived from natural casein.
  • Figure 5b represents the incorporation of [ H]-thymidine into cultured human bone marrow cells after 21 days incubation with or without (ctrl) 50 - 600 ⁇ g per ml peptides derived from natural casein. Bone marrow was donated by cancer patients in remission (BM Auto, closed squares, BM 1 , triangles, and BM 2,-open squares-) or healthy volunteers (BM normal, diamonds).
  • Figure 5c represents incorporation of [ H]-thymidine into cultured human Cord Blood cells after 14 days incubation with or without (ctrl) 50- 1000 ⁇ g per ml peptides derived from natural casein. Cord blood cells were donated by two separate donors (C.B. 1, triangles, C.B. 2, squares).
  • FIG. 6 shows a Table depicting the proliferation of blood cell progenitors from human bone marrow and cord blood in response to incubation with peptides derived from natural casein.
  • FIG. 7 shows a table depicting the effect of in-vitro incubation with Synthetic peptides derived from casein on the relative distribution of Megakaryocyte, Erythroid, Plasma and Dendritic cells (differential count) in CFU-GEMM colonies from murine bone marrow progenitor cells.
  • Cells were scored in the macroscopic colonies grown from murine bone marrow cells prepared similarly to the CFU-GEMM colonies previously described. Cells were incubated with hematopoietic factors, and 25 ⁇ g or more of Synthetic peptides derived from casein, for 14 days.
  • the differential count is expressed as the percentage of total cells represented by individual cell types.
  • FIG. 8 depicts the stimulation of peripheral white blood cell reconstitution in myeloablated, bone marrow transplanted mice in response to treatment with peptides derived from natural casein.
  • Cell counts represent the number of white blood cells (x 10 per ml, as counted in a haemocytometer).
  • Peptides, diamonds 1 mg per recipient human serum albumin
  • FIGs. lOa-lOf depict the penetration and nuclear uptake of FITC-conjugated peptides derived from natural casein in cultured human T-lymphocyte cells, as recorded by fluorescent microscopy.
  • FI and F2 are identical fractions of the FITC-conjugated peptides derived from natural casein.
  • Sup-Ti cells were incubated with 100 ⁇ g per ml FITC-conjugated peptides derived from natural casein as described in the Examples section that follows. At the indicated times, the cells were washed of free label, fixed in formalin and prepared for viewing and recording by Laser Scanning Confocal Microscopy.
  • Figures 10a through lOf are selected images of cells from consecutive incubation times, demonstrating FITC-conjugated peptides derived from natural casein penetrating the Sup-Ti cell membrane ( Figures 10a, 10b)- ⁇ and concentrating in the nucleus ( Figures 10c- lOf).
  • FIG. 1 1 shows a Table depicting the stimulation of Sup-Ti Lymphocyte cell proliferation in response to incubation with peptides derived from natural casein.
  • Sup-Ti cells (5000 per well) were incubated with increasing concentrations (50 - 1000 ⁇ g per ml) of peptides derived from natural casein, counted in their wells at the indicated times post culture and pulsed with
  • Proliferation index is the ratio of the average of
  • FIG. 12 shows a Table depicting inhibition of HIV-1 infection of CEM lymphocytes by peptides derived from natural casein.
  • CEM cells were either contacted with HIV-1 virus preincubated 3 hours with peptides derived from natural casein (3 hours), or preincubated themselves with increasing concentrations (50 - 1000 ⁇ g per ml) of peptides derived from natural casein for the indicated number of hours (24 and 48 hours) before contact with HIV-1 virus, as described in the Examples section that follows.
  • Control cultures were IF: CEM cells contacted with HIV-1 virus without pretreatment with peptides derived from natural casein, and UIF: CEM cells cultured under identical conditions without peptides derived from natural casein and without contact with HIV-1 virus.
  • FIG. 13 shows a Table depicting inhibition of HIV-1 infection of CEM lymphocytes by Synthetic peptides derived from casein.
  • CEM cells were contacted with HIV-1 virus which had been preincubated with various concentrations (10- 500 ⁇ g per ml) of synthetic peptides derived from natural casein (IP, 3P and 4P) for 3 hours (in the presence of the peptides), as described in the Examples section that follows.
  • IP, 3P and 4P synthetic peptides derived from natural casein
  • FIG. 14 depicts the prevention by peptides derived from natural casein of Type I (IDDM) Diabetes in female Non Obese Diabetic (NOD) mice.
  • IDDM Type I Diabetes
  • NOD Non Obese Diabetic mice.
  • Glucosuria was monitored at intervals during 365 days post treatment in female NOD mice receiving a once (triangles and squares) or twice weekly injection of 100 ⁇ g peptides derived from natural casein for 5 weeks (5 or 10 injections total) and untreated controls. All the controls developed glucosuria and subsequently died.
  • FIG. 15 depicts the reduction by Synthetic peptides derived from casein of diet-induced hypercholesterol/hyperlipidemia in female C57 Bl/6 mice.
  • Total cholesterol (TC), High Density (HDL) and Low Density Lipoproteins (LDL) were assayed in pooled blood of two (2) mice per sample from hypercholesterol/hyperlipidemic mice receiving (IP) casein-derived peptides B, C, 2a or 3P, or no treatment (control). "Normal" samples represent control mice not fed the atherogenic diet.
  • FIG. 16 shows a Table depicting the stimulation of hematopoiesis in cancer patients in response to injections of peptides derived from natural casein.
  • WBC White Blood Cells
  • PHT Platelets
  • RBC Erythrocytes
  • Hemoglobin gm per dl
  • Patient 1 relates to G.T.
  • patient 2 relates to E.C.
  • patient 3 relates to E.S.
  • patient 4 relates to J.R.
  • patient 5 relates to D.M.
  • FIG. 17 depicts the stimulation by peptides derived from natural casein of thrombocytopoiesis in a platelet-resistant patient with Acute Myeloid Leukemia (M-l).
  • Thrombocyte reconstitution was expressed as the change in platelet content of peripheral blood (PLT, x 10 per ml), counted as described above at the indicated intervals following intramuscular injection (as described in the Examples section that follows) of 100 ⁇ g peptides derived from natural casein.
  • FIG. 18 depicts the stimulation by peptides derived from natural casein of thrombocytopoiesis in a platelet-resistant patient with Acute Myeloid Leukemia (M-2).
  • Thrombocyte reconstitution was expressed as the change in platelet content of peripheral blood (PLT, x 10 per ml), counted as described above at the indicated intervals following intramuscular injection (as described in the Examples section that follows) of 100 mg peptides derived from natural casein.
  • the present invention is of biologically active peptides that are derived from or are similar to sequences identical with the N-terminus of the ⁇ Sl fraction of milk casein, compositions containing same and methods of utilizing same in, for example, stimulating and enhancing immune response, protecting against viral infection, normalizing serum cholesterol levels, and stimulating hematopoiesis.
  • the casein-derived peptides are non-toxic and can be used to treat and prevent, for example, immune pathologies, hypercholesterolemia, hematological disorders and viral-related diseases.
  • the term “treating” includes substantially inhibiting, slowing or reversing the progression of a disease, substantially ameliorating clinical symptoms of a disease.
  • the term “preventing” includes substantially preventing the appearance of clinical symptoms of a disease.
  • peptide includes native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides) and peptido-mimetics (typically, synthetically synthesized peptides), such as peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body.
  • Methods for preparing peptido-mimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, CA. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992), which is incorporated by reference as if fully set forth herein. Further detail in this respect are provided hereinunder.
  • a peptide according to the present invention can be a cyclic peptide.
  • Cyclization can be obtained, for example, through amide bond formation, e.g., by incorporating Glu, Asp, Lys, Orn, di-amino butyric (Dab) acid, di-aminopropionic (Dap) acid at various positions in the chain (-CO-NH or -NH-CO bonds).
  • Natural aromatic amino acids, Trp, Tyr and Phe may be substituted for synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
  • synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
  • Tables 1-2 below list all the naturally occurring amino acids (Table 1) and non-conventional or modified amino acids (Table 2).
  • a peptide according to the present invention can be used in a self standing form or be a part of moieties such as proteins and display moieties such as display bacteria and phages.
  • the peptides of the invention can also be chemically modified to give active dimers or multimers, in one polypeptide chain or covalently crosslinked chains.
  • a peptide according to the present invention includes at least two, optionally at least three, optionally at least four, optionally at least five, optionally at least six, optionally at least seven, optionally at least eight, optionally at least nine, optionally at least ten, optionally at least eleven, optionally at least twelve, optionally at least thirteen, optionally at least fourteen, optionally at least fifteen, optionally at least sixteen, optionally at least seventeen, optionally at least eighteen, optionally at least nineteen, optionally at least twenty, optionally at least twenty-one, optionally at least twenty-two, optionally at least twenty-three, optionally at least twenty-four, optionally at least twenty-five, optionally at least twenty-six, optionally between twenty-seven and sixty, or more amino acid residues (also referred to herein interchangeably as amino acids).
  • amino acid or “amino acids” is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phosphothreonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine.
  • amino acid includes both D- and L-amino acids.
  • ⁇ Sl casein refers to peptides as this term is defined herein, e.g., cleavage products of ⁇ Sl casein (referred to herein as peptides derived from natural casein), synthetic peptides chemically synthesized to correspond to the amino acid i o sequence of an N terminus portion of ⁇ S 1 casein (referred to herein as synthetic peptides derived from casein), peptides similar (homologous) to an N terminus portion of ⁇ Sl casein, for example, peptides characterized by one or more amino acid substitutions, such as, but not limited to, permissible substitutions, provided that at least 70 %, preferably at least 80 %, more preferably at least 90
  • ⁇ Sl casein refers to ⁇ Sl casein of a mammal, 0 including, but not limited to, livestock mammals (e.g., cow, sheep, goat, mare, camel, deer and buffalo) human beings and marine mammals.
  • livestock mammals e.g., cow, sheep, goat, mare, camel, deer and buffalo
  • NCBI GenBank Accession Nos.
  • CAA26982 (Ovis aries (sheep)), CAA51022 (Capra hircus (goat)), CAA42516 (Bos taurus (bovine)), 5 CAA55185 (Homo sapiens), CAA38717 (Sus scrofa (pig)), P091 15 (rabbit) and 097943 (Camelus dromedurius (camel)).
  • N terminus portion refers to M amino acids of ⁇ S 1 casein derived from the first 60 amino acids of ⁇ S 1 casein, wherein M is any of the integers between 2 and 60 (including the integers 2 and 60).
  • M is any of the integers between 2 and 60 (including the integers 2 and 60).
  • the term refers to the first M amino acids of ⁇ Sl casein.
  • the peptides of the invention can be obtained by extraction from milk as previously described, or by solid phase peptide synthesis, which is a standard method known to the man skilled in the art. Purification of the peptides of the invention is performed by standard techniques, known to the man skilled in the art, such as high performance liquid chromatography (HPLC). Milk casein fragmentation to obtain the peptides of the invention may be effected using various enzymatic and/or chemical means. As is further detailed hereinunder and exemplified in the Examples section that follows, the peptides of the present invention have a variety of therapeutic effects.
  • any of the peptides described herein can be administered per se or be formulated into a pharmaceutical composition which can be used for treating or preventing a disease.
  • a pharmaceutical composition includes as an active ingredient any of the peptides described herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition refers to a preparation of one or more of the peptides described herein, with other chemical components such as pharmaceutically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • the term "pharmaceutically acceptable carrier” refers to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • carriers examples, without limitations, of carriers are: propylene glycol, saline, emulsions and mixtures of organic solvents with water.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for formulation and administration of drugs may be found in
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, transdermal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active peptides into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the peptides of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer with or without organic solvents such as propylene glycol, polyethylene glycol.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer with or without organic solvents such as propylene glycol, polyethylene glycol.
  • organic solvents such as propylene glycol, polyethylene glycol.
  • penetrants are used in the formulation. Such penetrants are generally known in the art.
  • the peptides can be formulated readily by combining the active peptides with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the peptides of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active ingredient doses.
  • compositions which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active peptides may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the peptides according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the peptides described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active peptides may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the peptides to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water
  • the peptides of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • compositions herein described may also comprise suitable solid of gel phase carriers or excipients.
  • suitable solid of gel phase carriers or excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin and polymers such as polyethylene glycols.
  • a therapeutically effective amount also referred to as a therapeutically effective dose
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 or the IC 100 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data. Using these initial guidelines one having ordinary skill in the art could determine an effective dosage in humans.
  • toxicity and therapeutic efficacy of the peptides described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD 50 and the ED 50 .
  • the dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD 50 and ED 50 .
  • Peptides which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell cultures assays and animal studies can be used in formulating a dosage range that is not toxic for use in human.
  • the dosage of such peptides lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al., 1975, In: The Pharmacological Basis of Therapeutics, chapter 1, page 1).
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active ingredient which are sufficient to maintain therapeutic effect.
  • Usual patient dosages for oral administration range from about 1-1000 mg/kg/administration, commonly from about 10-500 mg/kg/administration, preferably from about 20-300 mg/kg/administration and most preferably from about 50-200 mg/kg/administration.
  • therapeutically effective serum levels will be achieved by administering multiple doses each day.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • dosing can also be a single administration of a slow release composition, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration.
  • Such notice for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • compositions comprising a peptide of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment or prevention of an indicated condition or induction of a desired event.
  • Suitable indica on the label may include treatment and/or prevention of an autoimmune disease or condition, viral disease, viral infection, bacterial infection, hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, hyperlipidemia, hypercholesterolemia, glucosuria, hyperglycemia, diabetes, AIDS, infection with HIV-1, helper T-cell disorders, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, hematopoietic stem cell proliferation, hematopoietic stem cell proliferation and differentiation, pre-leukemic conditions, leukemic conditions, immune system disorders resulting from chemotherapy or radiation therapy, and human immune system disorders resulting from treatment of diseases of
  • compositions according to the invention may be useful in maintaining and/or restoring blood system constituents, in balancing blood cell counts, in balancing levels of metabolites in the blood including sugar, cholesterol, calcium, uric acid, urea and enzymes such as alkaline phosphatase. Further, the pharmaceutical compositions of the invention may be useful in inducing blood cell proliferation, modulating white and/or red blood cell counts, particularly increasing white and/or red blood cell counts, elevating haemoglobin blood level and in modulating platelet counts.
  • balancing as used herein with relation to levels of certain physiological parameters, means changing the levels of referred parameters and bringing them closer to normal values.
  • normal values as used herein with relation to physiological parameters, means values which are in the range of values of healthy humans or animals.
  • the peptides of the invention balance counts of red blood cells, white blood cells, platelets and haemoglobin level.
  • the pharmaceutical compositions of the invention may be used for activating blood cell proliferation.
  • compositions may be used for the treatment and/or prevention of hemopoietic stem cell disorders, including platelet, lymphocyte, plasma cell and neutrophil disorders, as well as deficiency and malfunction in pre-leukemic and leukemic conditions and thrombocytopenia.
  • compositions may be used for the treatment and/or prevention of cell proliferative diseases.
  • pharmaceutical compositions of the invention are advantageous in the stimulation of the immune response during chemotherapy or radiation treatments, in alleviating the negative effects, reducing chemotherapy and irradiation-induced vomiting and promoting a faster recovery.
  • compositions of the invention may be used for the stimulation of human immune response during treatment of diseases associated with immune deficiency, for example HIV and autoimmune diseases.
  • compositions of the invention may also be intended for veterinary use.
  • the pharmaceutical compositions of the invention may be used in the treatment and/or prevention of, for example, disorders involving abnormal levels of blood cells, disorders involving hemopoietic stem cells production and differentiation, treatment of platelet, lymphocyte and/or neutrophil disorders, for the treatment of pre-leukemic and leukemic conditions and for the treatment of thrombocytopenia.
  • the pharmaceutical compositions of the invention may also be used in the treatment of cell proliferative diseases and diseases involving immune deficiency, such as HIV, and of autoimmune diseases.
  • the pharmaceutical compositions of the invention may be used for modulating the immune response during chemotherapy or radiation treatments, for example for reducing chemotherapy-associated vomiting.
  • the peptides of the invention exert a synergistic effect on human hematopoietic stem cell proliferation and differentiation with addition of other hematopoietic growth factors.
  • the potentiation of erythropoietin-mediated stimulation of erythroid colony formation and the dose-dependent enhancement of thrombopoietin (TPO) induction of megakaryocyte proliferation by peptides of the present invention was the potentiation of erythropoietin-mediated stimulation of erythroid colony formation and the dose-dependent enhancement of thrombopoietin (TPO) induction of megakaryocyte proliferation by peptides of the present invention.
  • TPO thrombopoietin
  • Recombinant human (rh) EPO is currently an approved therapy for indications such as renal anemia, anemia of prematurity, cancer- and AIDS-associated anemia, and for pre-elective surgical treatment (Sowade, B et al Int J Mol Med 1998;1 :305).
  • a method of treating an erythropoietin treatable condition the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of augmenting the effect of erythropoietin the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • Thrombopoietin is an early acting cytokine with important multilineage effects: TPO alone, or in combination with other early acting cytokines, can (i) promote viability and suppress apoptosis in progenitor cells; (ii) regulate hematopoietic stem cell production and function; (iii) trigger cell division of dormant multipotent cells; (iv) induce multilineage differentiation and (v) enhance formation of multilineage colonies containing granulocytes, erythrocytes, macrophages, and megakaryocytes (MK, CFU-GEMM).
  • TPO alone, or in combination with other early acting cytokines, can (i) promote viability and suppress apoptosis in progenitor cells; (ii) regulate hematopoietic stem cell production and function; (iii) trigger cell division of dormant multipotent cells; (iv) induce multilineage differentiation and (v) enhance formation of multilineage colonies containing
  • TPO stimulates the production of more limited progenitors for granulocyte/monocyte, megakaryocyte and erythroid colonies, stimulates adhesion of primitive human bone marrow and megakaryocytic cells to fibronectin and fibrinogen.
  • TPO is an important cytokine for clinical hematologists/transplanters: for the mobilization, amplification and ex vivo expansion of stem cells and committed precursor cells for autologous and allogeneic transplantation.
  • administration of TPO to healthy platelet donors has been employed to enhance pheresis yields.
  • clinical application of TPO therapy is complicated by, among other considerations, relatively high costs of the recombinant human cytokine rhTPO, and the potential antigenicity of TPO with repeated administration.
  • Combined treatment with TPO and the peptide of the present invention can provide inexpensive, proven non-toxic augmentation of TPOs effects on target cell proliferation and function.
  • the peptide of the present invention may be applied to the treatment of, in addition to the abovementioned conditions, disorders such as myelodysplastic syndrome (MDS), aplastic anemia and complications of liver failure.
  • MDS myelodysplastic syndrome
  • Pre-treatment of platelet donors with the peptide of the present invention, alone or in combination with TPO may even further enhance the efficiency of pheresis yields.
  • a method of treating a thrombopoietin treatable condition the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of augmenting the effect of thrombopoietin the method effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of enhancing peripheral stem cell mobilization the method effected by administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising effective amounts of thrombopoietin and a peptide derived from an N terminus portion of ⁇ SI casein.
  • a pharmaceutical composition for treating a thrombopoietin treatable condition comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for augmenting the effect of thrombopoietin comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for enhancing peripheral stem cell mobilization comprising, as active ingredients thrombopoietin and a peptide derived from an N terminus portion of ⁇ SI casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation and differentiation comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ S 1 casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing megakaryocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing erythropoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing Ieukocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing thrombocytopoiesis comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating thrombocytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a
  • a pharmaceutical composition for preventing or treating pancytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a i o pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating granulocytopenia comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein and a
  • a pharmaceutical composition for treating or preventing an indication selected from the group consisting of hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, dendrite cell deficiencies, 0 macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, myelodysplastic syndrome, aplastic anemia and bone marrow insufficiency, the pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion 5 of ⁇ S l casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising thrombopoietin and a purified peptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-25 and a pharmaceutically acceptable carrier.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating a donor of the donated blood stem cells with a peptide derived from an N terminus portion of ⁇ Sl casein and thrombopoietin prior to donation and implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of donated blood stem cells in a myeloablated recipient the method effected by treating the donated blood stem cells with a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin prior to implanting the donated blood stem cells in the recipient.
  • a method of enhancing colonization of blood stem cells in a myeloablated recipient comprising treating the blood stem cells with a peptide derived from an
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for treating a thrombopoietin treatable condition.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for augmenting the effect of thrombopoietin.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ S l casein, and a pharmaceutically acceptable carrier peptide derived from an N terminus portion of ⁇ Sl casein for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier enhancing colonization of blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for enhancing peripheral stem cell mobilization.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation and differentiation.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing megakaryocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing erythropoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing Ieukocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing thrombocytopoiesis.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating thrombocytopenia.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating pancytopenia.
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating granulocytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an indication selected from the group consisting of autoimmune disease or condition, viral disease, viral infection, hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, hyperlipidemia, hypercholesterolemia, glucosuria, hyperglycemia, diabetes, AIDS, HIV-1, helper T-cell disorders, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, immune system disorders resulting from chemotherapy or radiation therapy, human immune system disorders resulting from treatment of diseases of immune deficiency and bacterial infections.
  • an indication selected from the group consisting of autoimmune disease or condition, viral disease, viral infection, hematological disease, hematological deficiencies, thro
  • a pharmaceutical composition comprising, as active ingredients, thrombopoietin and a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an indication selected from the group consisting of hematological disease, hematological deficiencies, thrombocytopenia, pancytopenia, granulocytopenia, dendrite cell deficiencies, macrophage deficiencies, hematopoietic stem cell disorders including platelet, lymphocyte, plasma cell and neutrophil disorders, pre-leukemic conditions, leukemic conditions, myelodysplastic syndrome, aplastic anemia and bone marrow insufficiency.
  • a pharmaceutical composition comprising, as active ingredients, a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin, and a pharmaceutically acceptable carrier for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as active ingredients, a peptide derived from an N terminus portion of ⁇ S 1 casein and thrombopoietin, and a pharmaceutically acceptable carrier for enhancing colonization of blood stem cells in a myeloablated recipient.
  • the invention further relates to anti-bacterial pharmaceutical compositions comprising as active ingredient at least one peptide of the invention and to the use of the peptides of the invention as anti-bacterial agents.
  • peptides of the invention can be used in the treatment and prevention of blood cell disorders, cell proliferative diseases, diseases involving immune deficiency and autoimmune diseases.
  • a method of preventing or treating an autoimmune disease the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating a viral disease the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing viral infection is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing hematopoiesis the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of inducing hematopoietic stem cells proliferation is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing hematopoietic stem cells proliferation and differentiation is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing megakaryocytopoiesis the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of inducing erythropoiesis the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing Ieukocytopoiesis the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing thrombocytopoiesis the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing plasma cell proliferation is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing dendritic cell proliferation is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of inducing macrophage cell proliferation is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating thrombocytopenia is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating pancytopenia the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of preventing or treating granulocytopenia the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating hyperlipidemia is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating hypercholesterolemia is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating glucosuria is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ S 1 casein.
  • a method of preventing or treating diabetes is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating AIDS is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating infection by HIV the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a method of preventing or treating conditions associated with myeloablative doses of chemoradiotherapy supported by autologous bone marrow or peripheral blood stem cell transplantation (ASCT) or allogeneic bone marrow transplantation (BMT) the method is effected by administering to a subject in need thereof a therapeutically effective amount of a peptide derived from an N terminus portion of ⁇ Sl casein.
  • a pharmaceutical composition for preventing or treating an autoimmune disease comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating a viral disease comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing viral infection comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing hematopoietic stem cells proliferation and differentiation comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing megakaryocytopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing erythropoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing Ieukocytopoiesis the pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing thrombocytopoiesis comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing plasma cell proliferation the pharmaceutical composition comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing dendritic cell proliferation the pharmaceutical composition comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for inducing macrophage proliferation comprising, as an active ingredient a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating thrombocytopenia the pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating pancytopenia the pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating granulocytopenia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating hyperlipidemia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating hypercholesterolemia comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ S l casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating glucosuria comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating diabetes comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating AIDS comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for preventing or treating infection by HIV comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein and a pharmaceutically acceptable carrier.
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for preventing or treating a viral disease.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing viral infection. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for inducing hematopoiesis.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for inducing hematopoietic stem cells proliferation.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for inducing hematopoietic stem cells proliferation and differentiation. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for inducing megakaryocytopoiesis.
  • a peptide derived from an N terminus portion of ⁇ S 1 casein for inducing plasma cell proliferation. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for inducing dendritic cell proliferation. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for inducing macrophage proliferation.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating thrombocytopenia.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating pancytopenia. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating granulocytopenia.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating cholesteremia.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating diabetes. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating AIDS. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating infection by HIV.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for preventing or treating conditions associated with myeloablative doses of chemoradiotherapy supported by autologous bone marrow or peripheral blood stem cell transplantation (ASCT) or allogeneic bone marrow transplantation (BMT).
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a peptide derived from an N terminus portion of ⁇ Sl casein for treating a thrombopoietin treatable condition.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for augmenting the effect of thrombopoietin. Further according to the present invention there is disclosed the use of a peptide derived from an N terminus portion of ⁇ Sl casein for enhancing peripheral stem cell mobilization.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for enhancing colonization of donated blood stem cells in a myeloablated recipient.
  • a peptide derived from an N terminus portion of ⁇ Sl casein for enhancing colonization of blood stem cells in a myeloablated recipient.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating an autoimmune disease.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating a viral disease.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating a viral infection.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cell proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing hematopoietic stem cells proliferation and differentiation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing megakaryocytopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing erythropoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing Ieukocytopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing thrombocytopoiesis.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing plasma cell proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing dendritic cell proliferation. Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for inducing macrophage proliferation.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating thrombocytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating pancytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating granulocytopenia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide 5 derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating hyperlipidemia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically l o acceptable carrier for preventing or treating cholesteremia.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating glucosuria. 15 Further according to the present invention there is disclosed the use of a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating diabetes.
  • a 20 pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating AIDS.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide 25 derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating infection by HIV.
  • a pharmaceutical composition comprising, as an active ingredient, a peptide derived from an N terminus portion of ⁇ Sl casein, and a pharmaceutically acceptable carrier for preventing or treating conditions associated with myeloablative doses of chemoradiotherapy supported by autologous bone marrow or peripheral blood stem cell transplantation (ASCT) or allogeneic bone marrow transplantation (BMT).
  • ASCT autologous bone marrow or peripheral blood stem cell transplantation
  • BMT allogeneic bone marrow transplantation
  • a pharmaceutical composition comprising a purified peptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-25 and a pharmaceutically acceptable carrier.
  • the present invention successfully addresses the shortcomings of the presently known configurations by providing peptides for the treatment of human disease, which peptides are derived from the N terminus portion of ⁇ Sl casein and posses no detectable toxicity and high therapeutic efficacy.
  • HPLC analysis of peptides derived from natural casein Peptides derived from natural casein as described above were analyzed by HPLC in two stages. Initially, the lyophilized casein digests were separated using a C 18 reversed phase with a 0.1 % water triflouroacetic acid (w/w)-acetonitrile gradient. Detection was according to UV abso ⁇ tion at 214 nm. Following this the samples were analyzed by HPLC-Mass Spectroscopy (MS) equipped with an electrospray source. Mass calculations represent the mass of the ionized peptide samples, as derived from the retention times. Following separation, the amino acid composition of the peptides was determined with a gas-phase microsequencer (Applied Biosystems 470A).
  • Synthetic peptides derived from casein Peptides of increasing lengths corresponding to the N-terminal 2-26 amino acids of ⁇ Sl casein were synthesized by NoVetide Ltd., Haifa, Israel, with purity of >95 % (HPLC). Quality Control included: HPLC, Mass Spectrometry (El), Amino acid analysis and Peptide Content. Table 3 below provides the sequence of these peptides:
  • NOD mice are a commonly used model for research of autoimmune disease and human Juvenile Diabetes.
  • Six week old female NOD mice received either one or two injections per week of 100 ⁇ g of peptides derived from natural casein, for a total of 5 or 10 treatments.
  • Control mice received no treatment.
  • the severity of disease was determined according to glucosuria, which was measured using Combi test sticks [Gross, D.J. et al. (1994), Diabetology, 37:1195]. Results were expressed as the percent of glucosuria-free mice in each sample over a 365-day period.
  • Synthetic peptides derived from casein In another experiment, 6 week old female NOD mice received one injection per week of 100 ⁇ g of Synthetic peptides derived from casein for a total of 5 treatments. Control mice received no treatment. Results were expressed as the number of healthy mice in the various treated groups.
  • IPGTT Intraperitoneal Glucose Tolerance Test
  • NK cells Natural Killer (NK) cells: From human Peripheral Blood Stem Cells (PBSC): PBSC of G-CSF treated subjects were separated on a FICOLL gradient, washed twice with RPMI-1640 medium and seeded into 1.5 ml wells with or without peptides derived from natural casein or synthetic peptides derived from casein, as indicated, (0-500 ⁇ g per ml).
  • PBSC Human Peripheral Blood Stem Cells
  • effecto ⁇ target cell ratios of 50:1 and 100: 1, respectively in U-bottomed 96 well tissue culture plates.
  • the cells were incubated for 5 hours at 37 °C in 5 % C0 2 , 95 % air and precipitated by 5 minutes centrifugation at 1000 rpm.
  • 35 S release was measured in 50 ⁇ l samples of the supernatant liquid.
  • Bone marrow was collected from 4 untreated BALB/c and C57B1/6 mice. Bone marrow was harvested from the long bones of front and hind limbs of the mice by injection of medium using a 25 Gauge needle. Aspirated cells were washed with RPMI 1640, counted in a haemocytometer and vital-stained (20 ⁇ l of cells in 380 ⁇ l acetic acid/trypan blue), then seeded in culture bottles at 2-5 x 10 ⁇ cells per ml in RPMI-1640 containing 10 % Fetal Calf Serum, antibiotics and glutamine with or without 100 ⁇ g per ml peptides derived from natural casein.
  • NK activity is expressed as the percent radioactivity in the cell-free supernatants.
  • PB Peripheral blood
  • Affected patients received no treatment other than G-CSF supplementation prior to plasmapheresis.
  • Bone marrow (BM) cells were collected from consenting healthy patients or affected patients in remission following chemotherapy by aspiration.
  • Umbilical cord blood was collected during normal births.
  • Human cells of the various origins were separated on a FICOLL gradient, washed twice with RPMI-1640 medium, and seeded into 0.2 ml flat bottom tissue culture wells at the indicated concentrations with or without peptides derived from natural casein or with or without synthetic peptides derived from casein, as indicated. All treatments, including controls, were repeated in triplicate.
  • Inco ⁇ orated radioactivity was measured in a ⁇ scintillation counter.
  • Colon and K562 are established lines of cancer cells grown in culture. Both cell lines were grown in culture bottles in 5 % CO 2 , 95 % air at 37 °C, harvested and washed with medium before seeding in tissue culture wells at 4 x 10$ cells (K562) or 3 x 10- cells (Colon) per well. Peptides derived from natural casein were added to the wells, at the indicated concentrations, and after 9 (K562) or 3 (Colon) days of incubation labeled thymidine was added as described above. Harvesting and measurement of radioactive uptake was as described above.
  • PBSC Peripheral Blood Stem Cells
  • PBSC Peripheral Blood Stem cells from human subjects receiving G-CSF treatment were collected by plasmapheresis, separated on a FICOLL gradient, washed twice with RPMI-1640 medium containing 10 % Fetal Calf Serum and incubated in culture bottles at 37 °C in 5 % CO 2 , 95 % air with or without peptides derived from natural casein at the indicated concentrations.
  • T cells CD 3 surface antigen
  • NK cells CD 56 surface antigen
  • CD 3 fluorescent antibody CD TITC clone UHCTj
  • anti-CD56 fluorescent antibody CD 56 /RPE clone MOC-1
  • mouse IgGl/RPE and IgGl/FITC antibodies mouse IgGl/RPE and IgGl/FITC antibodies as a control.
  • FACS fluorescence activated cell sorting
  • CFU-GEMM multipotential colonies
  • the medium appropriate for the growth of multipotential colonies (CFU-GEMM), contained 1 % BSA (Sigma), 10" 4 M thioglycerol (Sigma), 2.8 x 10 ⁇ 4 M human transferrin (TF, Biological industries, Israel), 10 % WEHI-CM as a source of IL-3 and 2 units per ml erythropoietin (rhEPO, R & D Systems, Minneapolis). Colonies were scored after 8-9 days using an Olympus dark field microscope. They were picked with a micropipette, cytocentrifuged and stained with May-Grunwald-Giemsa for differential counts. At least 700 cells were counted for each preparation. Proliferation of Dendritic cells in CFU-GEMM: Multipotent
  • CFU-GEMM colonies grown from primary bone marrow cells as described for the assay of megakaryocyte proliferation above were collected, stained and counted for dendritic cells. At least 700 cells were counted for each preparation. Proliferation of Plasma cells in CFU-GEMM: Multipotent
  • CFU-GEMM colonies grown from primary bone marrow cells as described for the assay of megakaryocyte proliferation above were collected, stained and counted for plasma cells. At least 700 cells were counted for each preparation.
  • CFU-GEMM Multipotent (CFU-GEMM) colonies grown from primary bone marrow cells as described for the assay of megakaryocyte proliferation above were collected, stained and counted for macrophage cells. At least 700 cells were counted for each preparation.
  • CFU-GEMM Multipotent (CFU-GEMM) colonies grown from primary bone marrow cells as described for the assay of megakaryocyte proliferation above were collected, stained and counted for red blood cells. At least 700 cells were counted for each preparation.
  • CFU-GEMM Multipotent (CFU-GEMM) colonies grown from primary bone marrow cells as described for the assay of megakaryocyte proliferation above were collected, stained and counted for polymo ⁇ honuclear cells. At least 700 cells were counted for each preparation.
  • Proliferation of megakaryocyte- and erythroid forming cells from human bone marrow and cord blood cells A sample of bone marrow from an apparently healthy human being was processed by density gradient separation using Histopaque-107 (Sigma Diagnostics) to obtain a purified population of mononuclear cells (MNC).
  • Colony assays were performed in a plating medium containing final concentrations of 0.92 % methyl cellulose (4000 centripase powder, Sigma Diagnostic), rehydrated in Iscoves modified Dulbecco's medium containing 36 mM sodium bicarbonate (Gibco), 30 % fetal bovine serum (FBS) (Hyclone), 0.292 mg/ml glutamine, 100 units per ml penicillin and 0.01 mg per ml streptomycin (Biological Industries, Beit Haemek). Cord blood from normal births was collected and prepared as mentioned above.
  • Iscoves modified Dulbecco's medium containing 36 mM sodium bicarbonate (Gibco), 30 % fetal bovine serum (FBS) (Hyclone), 0.292 mg/ml glutamine, 100 units per ml penicillin and 0.01 mg per ml streptomycin (Biological Industries, Beit Haemek).
  • Cord blood from normal births was collected and prepared as mentioned
  • Colony assay medium containing 10 5 MNC per ml was plated in triplicate wells within a 24 well tissue culture plate (Greiner), 0.33 ml per well. The cultures were incubated at 37 °C in 5 % CO 2 , 95 % air and 55 % relative humidity with or without peptides derived from natural casein or synthetic peptides derived from casein, at the indicated concentrations. Plates were scored after 14 days for colonies containing more than 50 cells. Megakaryocytes were identified by indirect immunofluorescence using a highly specific rabbit antibody recognizing human platelet glycoproteins, and an FITC-conjugated goat anti-rabbit IgG.
  • Added growth factors included 15 ng per ml leucomax (GM-CSF) (Sandoz Pharma), and 5 % vol. per vol. human phyto-hemagglutinin-m (Difco Lab)-induced conditioned medium (CM) to induce development of granulocyte-monocyte colonies (CFU-GM). Erythropoietin (EPO) 2 units/ml was used to induce formation of erythroid colonies (burst-forming unit-erythroid-BFU-E).
  • GM-CSF leucomax
  • CM granulocyte-monocyte colonies
  • EPO Erythropoietin 2 units/ml was used to induce formation of erythroid colonies (burst-forming unit-erythroid-BFU-E).
  • human bone marrow cells from consenting volunteer donors or patients undergoing autologous bone marrow transplantation were precultured in medium containing 10-1000 ⁇ g per ml peptides derived from natural casein, grown in semi-solid agar, and scored for granulocyte-macrophage hematopoietic colonies (GM-CFU) at 7 or 14 days post treatment.
  • GM-CFU granulocyte-macrophage hematopoietic colonies
  • Megakaryocytopoiesis was measured in normal bone marrow cells from healthy consenting human donors by either scoring of the number of megakaryocytes in samples of liquid culture (RPMI-1640 plus 10 % human AB serum, glutamine and antibiotics) with or without 100 ⁇ g per ml peptides derived from natural casein, or in a methylcellulose assay for assessing colony formation. 2 x 10 ⁇ bone marrow cells were seeded in the presence of a standard growth factor combination with or without peptides derived from natural casein. In the methylcellulose assay megakaryocytes were counted with an inverted microscope on days 12-14 after seeding.
  • Clinical trials using peptides derived from natural casein In one series of trials, a single dose containing 50 mg peptides derived from natural casein was administered intra-muscular to human subjects in 3 depots, over a period of 2 hours. Clinical parameters were monitored at the indicated intervals. In other trials, patients at various stages of treatment for and/or remission from cancer and metastatic disease received peptides derived from natural casein once or twice, and were monitored for changes in the cell count of peripheral blood.
  • Peptides Peptides (either peptides derived from natural casein or synthetic peptides derived from casein (2-26 amino acids in length, see table 3) supplied as lyophilized powder were resuspended in RPMI complete medium and added to cell cultures at a final concentrations of 50 to 1000 ⁇ g per ml.
  • Cells Several types of freshly isolated human cells (primary cells) and cell lines are known to be susceptible to in vitro HIV-1 infection, although essentially any cell displaying even low surface levels of the CD 4molecule can be considered a potential target for HIV-1 infection. Two commonly used human cell lines which are highly sensitive for HIV-1 infection were chosen, CEM and Sup-Tl .
  • CEM is a human T4-lymphoblastoid cell line initially derived by G. E. Foley et al. [(1965), Cancer 18:522] from peripheral blood buffy coat of a 4-year old Caucasian female with acute lymphoblastic leukemia. These cells were continuously maintained in suspension in medium, and have been used widely for analysis of infectivity, antiviral agents and neutralizing antibodies.
  • Sup-Tl is a human T-lymphoblastoid cell line isolated from a pleural effusion of an 8-year old male with Non-Hodgkin's T-cell lymphoma [Smith, S. D. et al. [(1984) Cancer Research 44:5657]. This cell expresses high levels of surface CD4and is useful in studies of cell fusion, cytopathic effect and infectivity of HIV-1.
  • Sup-Tl cells are grown in suspension in enriched medium. Medium: Cells were grown in RPMI-1640 complete medium enriched with 10 % Fetal bovine serum, 2 mM glutamine and 2 mM penicillin- streptomycin (GIBCO).
  • HIV virus strain employed was HIV- HUB, originally designated HTLV-IIIB. Concentrated culture fluids of peripheral blood from several patients with AIDS or related diseases were used to establish a permanent productive infection in H-9 cells. This subtype B virus has high capacity to replicate in human T-cell lines. Viral titer was 5.38 ng per ml in stock solution.
  • FITC-labeled peptides FITC F-1300 (Fluorescein isothiocyanate, isomer I, Sigma (F25o-2) St. Louis, MI, USA) having excitation/emission maxima of about 494/520 nm, respectively, was employed.
  • the amine-reactive fluorescein derivative is probably the most common fluorescent derivatization reagent for covalently labeling proteins.
  • FITC-conjugated peptides derived from natural casein were prepared by covalent binding of FITC to the amine groups of lysine.
  • HIV-1 P 24 antigen capture assay An HIV-1 P 24 Antigen capture assay kit employed was designed to quantitate the HIV-1 P 24 core antigen, which is proportionally related to the degree of viral production in cells. This kit was purchased from the AIDS Vaccine program of the SAIC-NCI-Frederick Cancer Research Institute, P.O. Box B, Frederick, M.D 21702, USA and included 96 well plates coated with monoclonal antibody to HIV-1 P 24 , primary antibody-rabbit anti-HIV P 24 serum, secondary antibody-Goat anti-rabbit-IgG (H+L) peroxidase conjugated antibody, TMB peroxidase substrate system and lysed HIV-1 P 24 standard. The HIV-1 P 24 antigen capture assay was analyzed by Organon-Technica ELISA reader at 450 nm with a reference at 650 nm.
  • HIV-1 P 24 antigen capture ELISA HIV infection was measured with an indirect enzyme immunoassay which detects HIV-1 P 24 core antigens in tissue culture media. Tissue culture supernatant was reacted with primary rabbit anti-HIV- 1 P 24 antigen and visualized by peroxidase conjugated goat anti rabbit IgG. The reaction was terminated by adding 4N H SO_ ⁇ , wherein the intensity of the color developed is proportional to the amount of HIV-1 antigen present in the tissue culture supernatant.
  • Biological hazard level 3 (BL-3) laboratory All virus production isolation and infection, tissue culture of HIV-1 infected cells, P24 antigen containing supernatant harvesting andP 24 antigen capture ELISA, were performed in BL-3 facility of the Hebrew University, Hadassah Medical School and were in accordance with the bio safety practices set by the NIH and CDC (USA).
  • Flow cytometry A FACSort cell sorter (Becton & Dickinson, San Jose, CA. USA) was used to (i) determine the percentage of CD4 positive CEM and sup-Tl cells batches before infection with HIV-1 in order to assure the same degree of infection in each experiment; and (ii) detect T cells that harbor FITC conjugated peptides derived from natural casein in their cytoplasm and nuclei.
  • O2 incubator For viral culture production cells with HIV- 1 , cells and virus preheated with peptides derived from natural casein and cells which were further incubated with HIV-1, were all kept in humidified CO 2 incubator for the duration of the experiment.
  • HIV infection of human cultured CD4 cells For longer incubations, the cells (CEM, Sup-Tl) were preincubated with several increasing concentrations of peptides derived from natural casein (50-1000 ⁇ g per ml) or synthetic peptides derived from casein (10-500 ⁇ g per ml) for 24 (for synthetic and natural peptides) and 48 (only for natural peptides) hours and HIV- HUB (45 pg per ml final concentration) was added to each well thereafter. For the shorter incubations (3 hours), HIV- HUB was preincubated with the peptides for 3 hours and then added to cells (5000 cells/ well) in tissue culture plates.
  • Controls were IF (Infected, cells cultured with HIV-1 and without peptides), UIF (Uninfected, cells cultured without HIV-1 and without peptides) and UIF + Ch (Uninfected + peptides derived from natural casein, cells cultured in the presence of peptides derived from natural casein ⁇ 50-1000 ⁇ g per ml ⁇ ) to test the effect of peptides derived from natural casein and synthetic peptides derived from casein on cell viability and growth. Cells were counted for viability and proliferation rate on day 7, 10 and day 14 post infection (the day of P24 antigen culture supernatant harvest).
  • T cells were incubated with FITC conjugated peptides derived from natural casein in a 5 % CO 2 , 95 % air, 37 °C incubator, after which the cells were washed 3 times with phosphate buffer saline (PBS) to remove unbound FITC-peptides.
  • PBS phosphate buffer saline
  • HJ-thymidine incorporation test In order to test the effect of peptides derived from natural casein on T cell proliferation, several concentrations of peptides derived from natural casein (10 mg/ml stock in RPMI) were added to Sup-Tl cell cultures in 96 flat bottom microwell plate (5000 cells/well), as described for HIV-1 infection in Sup-Tl cells. Cells were counted and their viability was determined by trypan blue dye exclusion. They were pulsed with [ H]-thymidine at each time point (3, 7, 10 and 14 days) for 18 hours (over night) and harvested on glass fiber filters for radioactivity reading (Inco ⁇ oration of [ H]-thymidine into cellular DNA is proportional to degree of cell proliferation).
  • Toxicity of peptides derived from natural casein in normal, myeloablated and transplant recipient mice and guinea pigs Intramuscular, or intravenous injections of up to 5,000 mg peptides derived from natural casein per kg animal were administered in a single dose, or in three doses to normal animals. A variety of strains were employed, including BALB/c, C3H/HeJ and Non-Obese Diabetic (NOD) mice. The mice were either monitored for 10 months before sacrifice and post-mortem examination (toxicity assay) or observed for 200 days (survival rate). Guinea pigs received a single intramuscular injection of 20 mg peptides derived from natural casein per animal.
  • mice were sub-lethally irradiated at a source to skin distance of 70 cm, dosage of 50 cGy per minute, for a total of 600 cGy.
  • the irradiated mice were reconstituted with syngeneic bone marrow as described above and injected intravenously 24 hours later with 1 mg per animal peptides derived from natural casein, synthetic peptides derived from casein (13-26 amino acids, see Table 3 above), or human serum albumin (controls), following a double-blinded protocol.
  • Leukocyte reconstitution was determined according to cell count in peripheral blood collected at indicated intervals from 6 to 12 days post treatment. Platelet reconstitution was determined by cell count in blood collected from the retro orbital plexus, into EDTA-containing vials, at indicated intervals from day 6 to day 15 post treatment.
  • CBA mice were lethally irradiated (900 cGy), reconstituted with BM cells and treated with peptides derived from natural casein or human serum albumin as described above. Platelet reconstitution was assayed as mentioned above.
  • mice were irradiated (800 cGy), reconstituted and injected intraperitoneally with 100 ⁇ g synthetic peptides derived from casein (peptides 3 a and 4P, representing the first 6 and 12 amino acids of the N terminus of ⁇ Sl casein, respectively - see Table 3 above) daily, on days 4, 5, 6 and 7 post-transplantation. Platelet reconstitution was assayed at 10 and 12 days post-transplantation.
  • casein peptides 3 a and 4P, representing the first 6 and 12 amino acids of the N terminus of ⁇ Sl casein, respectively - see Table 3 above
  • mice C57B1/6 mice were lethally irradiated at a source to skin distance of 70 cm, dosage of 50 cGy per minute, for a total of 900 cGy.
  • the irradiated mice were reconstituted with syngeneic bone marrow cells from mice which were either treated a day prior to bone marrow collection with 1 mg per animal peptides derived from natural casein or with saline (controls), following a double-blinded protocol. In one experiment mice survival was monitored for 18 days. In another experiment mice were sacrificed after 8 days and spleen colonization monitored.
  • Synthetic peptides derived from casein significantly reduce Cholestrol levels The ability of synthetic caesin derived peptides to reduce cholesterol levels in 7-week old female C57B1/6J mice was assessed after feeding an atherogenic diet. The mice were divided into groups of 8. One control group was fed a normal diet. A second control group was fed the modified Thomas Hartroft diet containing cholate (#TD 88051 : Teklad, Madison, WI) [Gerber, D. W. et al., Journal of Lipid Research. 42, 2001]. The remaining experimental groups were all fed the modified Thomas Hartroft diet.
  • Peptides derived from natural casein Originating from the observation that curdled milk occasionally failed to support bacterial growth, a casein fragment possessing bacteriocidal properties was isolated from milk proteins (United States Patent No. 3,764,670 to Katzirkatchalsky, et al). Crude peptides derived by proteolysis of natural casein were prepared by acid precipitation of the soluble fraction of the casein proteolytic digest, dialysis and lyophilization. When tested for biological activity after extended storage, it was noted that this crude preparation, when lyophilized and stored at 4 °C, remained active (in vitro and in vivo) for at least 24 months.
  • a major component of the crude peptides derived from natural casein preparation is the N-terminal fragment of ⁇ S 1 casein.
  • Peptides derived from natural casein are non-toxic in rodents and humans: Extensive investigation of the short and long term effects of high doses of peptides derived from natural casein on mice, rats, guinea pigs and human volunteers confirmed the absence of toxicity, teratogenicity or adverse side effects of the preparation. In one series of tests, single doses representing 7,000 times the estimated effective dose of peptides derived from natural casein were administered intra muscularly to mice. Standard post-mortem pathology examination of the mice at 14 days post treatment revealed no toxic effects on internal organs or other abnormalities.
  • peptides derived from natural casein were safe when administered to humans as well. Comparison of blood and urine samples from seven healthy human volunteers before, during and 7 days after intramuscular injection of peptides derived from natural casein revealed no changes in any of the clinical parameters. No other negative effects were observed.
  • C57B1/6 mice were lethally irradiated and reconstituted with syngeneic bone marrow from mice that were either treated a day prior to bone marrow collection with 1 mg per animal peptides derived from natural casein or not so treated, survival of irradiated mice that received bone marrow from treated mice far exceeded that of irradiated mice that received bone marrow from non treated mice (survival of irradiated mice that received bone marrow from treated mice was 15 out of 18, 10 days post irradiation; whereas survival of irradiated mice that received bone marrow cells from saline-treated control mice was 4 out of 17, 10 days post irradiation).
  • Spleens derived from irradiated mice that received bone marrow from treated mice included about twice to three times as many colonies per spleen, as compared to spleens of irradiated mice that received bone marrow cells from saline-treated control mice (1-5 colonies as compared to 0-3 colonies).
  • NK Natural killer
  • cytotoxic T cells are crucial to the immune system's ability to protect against invasion by both infectious pathogens and cancer cells, by both active cytotoxicity and the secretion of immunoregulatory lymphokines. Immune compromise, such as in AIDS or following chemotherapy, results in abnormal, weakened T or NK cell activity.
  • NK natural killer
  • cytotoxic T cells are crucial to the immune system's ability to protect against invasion by both infectious pathogens and cancer cells, by both active cytotoxicity and the secretion of immunoregulatory lymphokines.
  • Immune compromise such as in AIDS or following chemotherapy, results in abnormal, weakened T or NK cell activity.
  • normal murine bone marrow cells from BALB/c and C57B1/6 mice were cultured in the presence of 100 ⁇ g per ml peptides derived from natural casein, a clear increase in NK activity was observed in both effecto ⁇ target cell ratio groups. Moreover, comparison between the two groups revealed a clear dose response relationship.
  • NK activity was measured in blood samples taken from one patient and incubated at two effecto ⁇ target cell ratios with increasing peptides derived from natural casein concentration. Only 4 % 35 S release was measured in the control, untreated PBSC culture. Almost the same percent radioactivity (4 %) was found at the lowest peptide concentration (5 ⁇ g per ml). However, at higher peptide concentrations, in the range of lO ⁇ g per ml up to lOO ⁇ g per ml, a release of 10.8-14.9 % 35 S was measured for effecto ⁇ target cell ratios of 100: 1 and 8.3-14.5 % 35 S for effector target cell ratios of 50:1 ( Figure 2a).
  • CD56 surface antigen positive (NK) cells In another series of experiments Peripheral Blood Stem Cells (PBSC) from 5 human donors receiving GCSF treatment were incubated with peptides derived from natural casein for 10, 14, or 28 days, then assayed for presence of the CD 56 antigen. A sometimes dramatic increase in CD 56 antigen detection was observed in the peptide-treated cells from all the donors but one (patient 1). A representative response is depicted in Figure 3a: Following 10 days of incubation with or without peptides derived from natural casein, the presence ofCD56 surface antigen-positive (NK) cells was detected by direct immunofluorescent staining. Overall, incubation with peptides derived from natural casein increased the mean percentage of the cells positively stained for CD56 from 0.64 % in the control group to 2.0 % following treatment ( Figure 3a).
  • PBSC Peripheral Blood Stem Cells
  • Peptides derived from natural casein stimulate the proliferation of CD3 surface antigen-positive (T) cells The effect of peptides derived from natural casein on the proliferation of CD3 surface antigen-positive (T) cells in PBS cells from 5 subjects was assayed by direct immunofluorescence. In all but one patient (patient 4), 14 days incubation with peptides derived from natural casein significantly increased T-cell proliferation, up to more than 5 fold in some. Taken together, the mean percentage of the cells positively stained for CD3 increased from 19.45 % in the control group to 35.54 % in the treated group ( Figure 3b).
  • Peptides derived from natural casein stimulate the proliferation of - CD56 and CD3 (NK/ T-cells) positive cells:
  • PBSCs from 7 patients were incubated with peptides derived from natural casein for 28 days, and the effect on proliferation of NK/T cells (CD56 and CD3 surface antigen-positive) was detected by direct immunofluoresence.
  • T-cell greater than 5 fold in some cases (patient 6), while the mean percentage of the CD3- positive (T-) cells increased from 2.08 % in the control group to 6.49 % in the treated group.
  • peptides derived from natural casein stimulate the proliferation of both T-lymphocytes and Natural Killer cells from normal murine and human blood cell progenitors.
  • the greatest immune-stimulatory effect of the peptides derived from natural casein was noted in human donors having initially low T- and NK cell levels ( Figures. 3a-c).
  • Synthetic peptides derived from casein stimulate human lymphocyte proliferation in vitro When synthetic peptides derived from casein representing the first 3 to 26 residues of ⁇ S 1 casein were incubated with human PBSC cells from healthy and cancer patients (see below), a significant increase in NK cell activity was observed. Target cell lysis was greatest (from 3 to greater than 5 fold that of controls) in Non-FIodgkin's Lymphoma and Breast Cancer patient's PBSC cultures after two days incubation with as little as 10 ⁇ g per ml of peptides containing the first 9 or more residues of ⁇ Sl casein ( Figure 4).
  • NK cell activity Similar stimulation of NK cell activity was observed when PBS cells from human donors with hematopoietic disease were incubated with Synthetic peptides derived from casein representing the first 3 amino acid residues of ⁇ S 1 casein. Incubation of the PBS cells with the peptides increased target cell lysis from 2- to greater then 8- fold that of the untreated controls. Of the 5 patients tested, three (3) responded to 25 ⁇ g/ml peptide concentration, one (1) responded to 100 ⁇ g/ml peptide concentration and one (1) to 250 ⁇ g/ml. Three out of the five (5) patients responded at 25 ⁇ g/ml..
  • Blood cell progenitors differentiate into a variety of blood cells: macrophages, monocytes, granulocytes, lymphocytes, erythrocytes and megakaryocytes. Progenitor cells are abundant in bone marrow, but are also found in peripheral blood after Granulocyte Colony Stimulating Factor treatment (PBSC cells), and fresh Cord Blood.
  • PBSC cells Granulocyte Colony Stimulating Factor treatment
  • FIG. 5a-5c When increasing concentrations (50-600 ⁇ g per ml) of peptides derived from natural casein were added to cultures of human Bone Marrow, PBSC and Cord Blood, an increase in cell proliferation, as measured by [ H]-fhymidine inco ⁇ oration was noted ( Figures 5a-5c).
  • Human PBSC proliferation was most greatly effected by 300 ⁇ g per ml (Figure 5a) after 15 days in culture. An even greater effect was noted for Cord Blood cells in culture (3 to 4 fold increase in [ H]-thymidine inco ⁇ oration) after 14 days incubation (but not after 7 days) with peptides derived from natural casein (600 ⁇ g per ml, Figure 5c). Cultured human bone marrow cells from three out of four donors also reacted strongly (3 to 5 fold increase in inco ⁇ oration) to peptides derived from natural casein (300 ⁇ g per ml) after 21 days incubation (Figure 5b).
  • peptides derived from natural casein stimulate proliferation of human blood cell progenitors from bone marrow as well as other sources.
  • incubation of cultured human K562 (Chronic Myeloid Leukemia) and Colon (Colon cancer) cell lines with high concentrations (up to 500 ⁇ g per ml) of peptides derived from natural casein under similar conditions had no effect on [ 3 H]-thymidine inco ⁇ oration.
  • peptides derived from natural casein stimulate proliferation of human blood cell progenitors but not growth of cancerous cells in vitro.
  • CFU-GM Granulocyte and Monocyte
  • CFU-GEMM Granulocyte, Erythroid, Macrophage and Megakaryocyte
  • synthetic peptides derived from casein representing the first 5 to 24 amino acids of ⁇ Sl casein increase the percentage of early and late megakaryocytes from 15 % without the synthetic peptide to more than 40 % with 25 ⁇ g per ml of synthetic peptides (Figure 7).
  • synthetic casein derived peptides representing the first 5, 6, 1 1, 12, 17, 18, 19, 20, 21 and 24 amino acids stimulated a significant increase in megakaryocyte formation and development in primary murine bone marrow culture..
  • Somewhat milder, yet appreciable, stimulation was observed with the other synthetic peptides derived from casein.
  • Peptides derived from natural casein stimulate Megakaryocytopoiesis in cultured human bone marrow cells: When 100 ⁇ g per ml peptides derived from natural casein were added under similar conditions to human bone marrow cell cultures from healthy donors, CFU-GM colony formation was increased with or without additional stimulating factors (GM-CSF, CM). Peptides derived from natural casein also stimulated erythroid cell forming colonies in the presence of erythropoietin. Treatment of the human bone marrow cells with thrombopoietin (TPO) stimulates megakaryocyte (MK) colony formation.
  • TPO thrombopoietin
  • the relative cell number counts in the cultured human bone marrow and cord blood colonies reflect megakaryocyte cell proliferation in response to addition of 25 ⁇ g per ml peptides derived from natural casein (see Table shown in Figure 6).
  • incubation of cultured human primary bone marrow and cord blood cells with peptides derived from natural casein stimulates the development and proliferation of both committed megakaryocyte and erythroid cell colonies.
  • Peptides derived from natural casein and synthetic peptides derived from natural casein potentiate the effect of Erythropoietin (EPO) in cultured human bone marrow cells: The effect of natural and synthetic peptides derived from casein on erythroid cell proliferation in cultured human bone marrow cells was assessed under the same conditions outlined hereinabove for megakaryocytopoiesis.
  • peptides derived from natural casein When added in the presence of EPO, 50 -300 ⁇ g/ml peptides derived from natural casein, or 100 ⁇ g/ml Synthetic peptides derived from casein (F, Table 3, SEQ ID NO: 18) stimulated a one and one-half (synthetic peptide) to four-fold proliferation of erythroid cell precursors (appearance of BFU-E colonies) compared to the bone marrow cells treated with EPO alone.
  • peptides derived from natural casein and synthetic derivatives thereof act to potentiate the erythropoietic-stimulating effects of EPO, and as such can be used to augment of a wide range of clinically important EPO-mediated effects.
  • Synthetic peptides derived from casein stimulate Dendritic cells proliferation in murine CFU-GEMM The effect of Synthetic peptides derived from casein on dendritic cell proliferation in murine primary bone marrow cells was assessed under the same conditions outlined for the stimulation of megakaryocytes.
  • SI casein stimulated the proliferation of dendritic cells, from 2.2 % and up to 23 % of total cells compared with 0.1 - 0.2 % dendritic cells in the cell samples incubated without Synthetic peptides derived from casein ( Figure 7).
  • Synthetic peptides derived from casein stimulate Plasma cell proliferation in murine CFU-GEMM The effect of Synthetic peptides derived from casein on plasma cell proliferation in murine primary bone marrow cells was demonstrated under the same conditions outlined for the stimulation of megakaryocytes.
  • Synthetic peptides derived from casein representing the first: 2, 3, 5, 7, 1 1, 16, 17, 18, 19, 20, 21, 22, 23 and 24 and 26 amino acids of ⁇ Sl casein significantly stimulated the proliferation of plasma cells, from 1.5 % and up 12.3 % of total cell count, compared with 0.3 % of total without Synthetic peptides derived from casein (Figure 7).
  • Synthetic peptides derived from casein stimulate Macrophage proliferation in CFU-GEMM The effect of Synthetic peptides derived from casein on macrophage proliferation in murine primary bone marrow cells was demonstrated under the same conditions outlined for the stimulation of megakaryocytes. Incubation of cells with synthetic peptides derived from casein representing the first: 7, 9, 16, and 23 amino acids of ⁇ Sl casein significantly stimulated the proliferation of macrophages, from approximately 17 % of total cell count in controls, to nearly 30 % of total in cells incubated with Synthetic peptides derived from casein (Figure 7).
  • Synthetic peptides derived from casein stimulate Red Blood Cells proliferation in CFU-GEMM The effect of Synthetic peptides derived from casein on red blood cell proliferation in murine primary bone marrow cells was demonstrated under the same conditions outlined for the stimulation of megakaryocytes. Incubation of cells with Synthetic peptides derived from casein representing the first 4 amino acids from the N terminus of ⁇ Sl casein significantly stimulated the proliferation of red blood cells, from 53 % of total cell count in controls, to 71 % of total in cells incubated with the synthetic peptide derived from casein ( Figure 7). Synthetic peptides derived from casein stimulate Polymorphonuclear
  • Peptides derived from natural casein stimulate hematopoiesis in vivo following irradiation and bone marrow transplant Myeloablative therapy may lead to life-threatening reduction in thrombocytes and leukocytes, which may persist despite administration of blood cells and growth factors. The following demonstrates the effect of peptides derived from natural casein following irradiation and bone marrow transplantation.
  • Peptides derived from natural casein enhance leukocyte and platelet reconstitution following syngeneic bone marrow transplantation in mice:
  • sub-lethally irradiated (600 cGy) minimally bone marrow-reconstituted
  • mice treated with the peptides derived from natural casein demonstrating a significant increase over the human serum albumin-treated controls which became even more pronounced by day 15 ( Figure 9).
  • peptides derived from natural casein enhance platelet and leukocyte reconstitution following transplantation with limiting numbers of bone marrow cells. It is expected that this effect will be further increased in reconstitution with optimal, rather than limiting numbers of bone marrow cells.
  • Synthetic peptides derived from casein enhance platelet reconstitution following syngeneic bone marrow transplantation in mice In order to confirm the observed ability of synthetic peptides derived from casein to enhance megakaryocyte proliferation in hematopoietic stem cell cultures (see Figures 6 and 7), the peptides' effects on platelet reconstitution in vivo was investigated.
  • Treatment with peptide 4P increased counts by 29 % (872 X 10 3 /ml compared with 676 X 10 3 /ml in the control group) at 12 days post transplantation while treatment with peptide 3a increased counts by up to 35.5 % (229 X 10 3 /ml compared with 169 X 10 3 /ml in the control group) at 10 days, and up to 13.5 % (622 X 10 /ml compared with 461 X 10 3 /ml in the control group) at 12 days post transplantation.
  • the same synthetic peptides derived from casein enhance megakaryocyte proliferation in vitro and platelet reconstitution following bone marrow transplantation in vivo.
  • Peptides derived from natural casein inhibit in vitro infection of lymphocytic T cell lines by HIV-1 virus Penetration of peptides derived from natural casein into lymphocytic
  • Tcells In order to investigate the mechanisms of immune stimulatory and anti-viral effects of peptides derived from natural casein, susceptible Sup-Tl and CEM cultured human T-cells were treated with peptides derived from natural casein prior to in vitro infection with HIV-1 virus. Fluorescent microscopy revealed that FITC-conjugated peptides derived from natural casein (100 ⁇ g per ml) penetrated the Sup-Tl cells when incubated therewith as described above ( Figures 10a- f). A small amount of label was observed in the cytoplasm of the cells after 15 minutes ( Figures lOa-b). At 30 minutes ( Figures lOc-d) more label was observed in the cytoplasm, with limited nuclear uptake.
  • Synthetic peptides derived from casein enhance human lymphocyte proliferation The presence of synthetic peptides derived from casein (all peptides listed in Table 3) in the culture medium resulted in increased Sup-Tl cell counts over a period of 10 days. The increase was similar for all synthetic peptides. The greatest increases in lymphocyte cell number in infected cells were observed for 250 ⁇ g and 500 ⁇ g per ml of peptide representing the first 9 amino acids (80 % and 33 %, respectively) (data not shown).
  • Peptides derived from natural casein inhibit HIV-1 infection in human lymphocyte cells: Susceptible CEM lymphocyte cells pretreated with peptides derived from natural casein (50-1000 ⁇ g per ml) 24 or 48 hours prior to incubation with HIV-1, or exposed to HIV-1 pretreated 3 hours with peptides from natural casein, exhibited enhanced cell proliferation and reduced levels of viral infection compared to untreated controls.
  • peptides derived from natural casein penetrate human cultured lymphocyte cells and their nuclei, enhance cell growth, and significantly reduce the susceptibility of CD4 cells to HIV-1 infection.
  • peptides derived from natural casein are expected to be useful both at preventing HIV infection and for post infection treatment of HIV infected and AIDS patients.
  • Synthetic peptides derived from casein inhibit HIV-1 infection in human lymphocyte cells The ability of Synthetic peptides derived from casein to inhibit HIV-1 infection in human lymphocyte cells was demonstrated using CEM-lymphocyte cells under the same conditions outlined above. Three hours pretreatment of CEM lymphocytes with the synthetic peptide derived from casein representing the first 3 amino acids of ⁇ S 1 casein conferred a significant degree of resistance to infection following incubation with HIV-1. Lymphocyte cell numbers were 1.29 x 10 6 (lOO ⁇ g per ml) and 2.01 x 10 6 (500 ⁇ g per ml) in the treated cells as compared to the infected HIV-1 control of 1.06 x 10 6 ( Figure 13).
  • HIV-1 infection levels in the same cells were significantly reduced in the peptide treated cells (0.17 and 0.14ng P 24 Antigen/ml withlOO ⁇ g/ml and 500 ⁇ g/ml respectively), as compared to the untreated controls (0.52 ng P 24 Ag/ ml). Likewise, significant inhibition of HIV-1 infection was observed in the peptide treated cells (0.17 and 0.14ng P 24 Antigen/ml withlOO ⁇ g/ml and 500 ⁇ g/ml respectively), as compared to the untreated controls (0.52 ng P 24 Ag/ ml). Likewise, significant inhibition of HIV-1 infection was observed in the
  • HIV- P 24 antigen assay at 7 days post infection revealed significant reduction in HIV-1 infection levels in treated cultures (0.26 and 0.18ng P 24 Ag per ml for 10 and 25 ⁇ g per ml respectively, as compared to the control of 0.52 ng P 24 Ag per ml).
  • N-terminal amino acids of ⁇ S-1 casein N-terminal amino acids of ⁇ S-1 casein.
  • Non-Obese Diabetic mice Non-Obese Diabetic mice spontaneously develop Juvenile (Type I, IDDM) Diabetes, an autoimmune condition causing inflammation of the pancreatic ⁇ cells and ending in disease and death.
  • Female NOD mice are extremely susceptible, demonstrating evidence of macrophage invasion of the pancreatic islet interstitial matrix as early as 5 weeks old.
  • a once or twice weekly injection of 100 ⁇ g peptides derived from natural casein for 5 weeks (5 or 10 injections total) were completely effective in preventing the glucosuria associated with the onset and course of the disease.
  • Plasma glucose levels were determined with a
  • Glucose Analyzer 2 (Beckman Instruments.Fullerton, CA) and expressed as mmol/L.
  • IPGTT performed with 6 healthy female control mice: 0 min- 110 mmol/L;
  • IPGT glucose tolerance
  • Synthetic casein- derived peptides significantly reduce Total Cholestrol blood levels (TC), Low Density Lipoprotein (LDL) and High Density Lipoprotein (HDL): Intraperitoneal administration of Synthetic peptides derived from casein caused a significant reduction in the blood lipid (HDL, LDL and TC) values in experimentally hypercholesterolemic mice. After one week of the atherogenic Thomas Hartroft diet, the blood cholesterol levels of the mice had risen to the levels of 318mg/dl.
  • Clinical trials with peptides derived from natural casein Patients received intramuscular injections of 50 mg peptides derived from natural casein each, divided into three depots, as indicated.
  • Peptides derived from natural casein stimulates hematopoiesis in cancer patients The hematology profiles of six cancer patients who had received or were receiving chemotherapy were examined before and following administration of peptides derived from natural casein, as indicated. Special attention was paid to changes in the Platelet (PLT), Leukocyte (WBC), Erythrocyte (RBC) and Hemoglobin (HGB) values, representing thrombocytopoiesis, Ieukocytopoiesis, and erythrocytopoiesis, respectively.
  • PLT Platelet
  • WBC Leukocyte
  • RBC Erythrocyte
  • HGB Hemoglobin
  • G.T. Male patient, Patient 1: Patient had ovarian cancer, undergone a hysterectomy followed by chemotherapy. She received two intramuscular injections of peptides derived from natural casein at two and then two and one half months post operation. No chemotherapy was administered between the first and second administrations of peptides derived from natural casein. Blood tests from 6 days post first injection, 7, and 13 days post second injection reflect a considerable increase in platelet and WBC components, as well as increased RBC ( Figure 16).
  • E.C. Male patient, Patient 2: Patient underwent a radical mastectomy for lobular carcinoma in 1983, and six years later suffered from gastric metastases. Three days prior to commencement of chemotherapy, she received one intramuscular injection (in three depots) of peptides derived from natural casein by injection, and a second 10 days after the chemotherapy. Although the blood counts from 10 and 16 days post chemotherapy indicated an attenuation of the depressed hematological profile usually encountered following chemotherapy, the most significant effects of peptides derived from natural casein were noted 3 days after the first injection, prior to the chemotherapy ( Figure 16).
  • E.S. Male patient, Patient 3: Patient was suffering from widespread metastatic dissemination of a breast carcinoma first discovered in 1987. Two years later, she received a first intramuscular injection of peptides derived from natural casein, and a second 23 days later. No additional therapy was administered during this period. Blood tests indicate a strong enhancement of PLT seven days after the first treatment and a significant increase in RBC and WBC seven days after the second treatment (Figure 16).
  • J.R. (Female patient, Patient 4): Patient's diagnosis is breast cancer with bone metastases. She received one intramuscular injection of peptides derived from natural casein 8 days before commencing chemotherapy, and another, 14 days later. The most significant effect is clearly seen in the rapid return of WBC levels following chemotherapy-induced depression ( Figure 16).
  • D.M. (Female patient, Patient 5): Patient suffering from hepatic cancer with widespread metastatic dissemination. She received three intramuscular injections of peptides derived from natural casein at 10, 8 and 6 days before receiving chemotherapy. A second series of injections was initiated 10, 12 and 14 days following the chemotherapy treatment.
  • administration of peptides derived from natural casein to cancer patients results in improved hematological profiles, specifically enhanced erythropoiesis, Ieukocytopoiesis and thrombocytopoiesis, and is capable of moderating and shortening the duration of chemotherapy-induced depression of blood components.
  • Peptides derived from natural casein stimulates thrombocytopoiesis in transplant recipients with resistant thrombocytopenia: Prolonged transfusion-resistant thrombocytopenia with episodes of severe bleeding, may be a life threatening complication of bone marrow transplantation, especially where traditional therapies are ineffective. Two patients with severe resistant thrombocytopenia were treated with peptides derived from natural casein.
  • M-l Female patient: 32 year old patient suffering from Acute
  • Myeloid Leukemia in complete remission following autologous stem cell transplantation. She had experienced two life-threatening bleeding episodes, involving pulmonary hemorrhage and a large obstructive hematoma in the soft palate. At more than 1 14 days post transplantation, platelet counts were refractive to rhIL-3, rhIL-6, intravenous gamma globulin, and recombinant erythropoietin. Following two intra muscular treatments of 50 mg peptides derived from natural casein (each treatment divided into three depots), her condition improved immediately. Along with the rapid return of normal platelet counts (Figure 17), her distal limb bleeding with exertion and patechyae subsided, she was able to resume walking, and returned to her home overseas with no complications or side effects.
  • M-2 (Male patient): 30 year old patient suffering from Acute Myeloid Leukemia in a second complete remission following autologous stem cell transplantation, exhibiting totally resistant platelet counts and massive gastrointestinal bleeding episodes. He required daily transfusions of packed cells, had developed hypoalbuminia, and failed to respond to extensive therapy with rhIL-3, rhIL-6 and gamma globulin. Following two intramuscular treatments, each of 50 mg peptides derived from natural casein in three depots 86 days post transplantation, rapid platelet reconstitution (Figure 18) and gradual discontinuation of the bleeding was observed. No further treatment was required, and the patient is presently completely asymptomatic with normal platelet count.
  • Peptides derived from natural casein decreases triglycerides and Total Cholesterol in familial hyperlipidemia: M.S. (Female patient): Patient is a 38 year old female with family history of hyperlipidemia. Before treatment with peptides derived from natural casein, blood chemistry profile revealed elevated total cholesterol (321 mg per dl), triglycerides (213 mg per dl; normal range 45 - 185 mg per dl) and elevated LDL-cholesterol (236.4 mg per dl; normal range 75 - 174 mg per dl).
  • D. G. (Male patient): Patient is a 75 year old male suffering from anemia and hypoglobinemia (depressed RBC, HGB, HCT, MCH and MCHC) associated with extensive occult bleeding. One month after receiving one intramuscular injection of 50 mg peptides derived from natural casein (in three depots), a significant reduction of the anemia was observed. After two months, RBC approached normal values (4.32 instead of 3.44 M per ⁇ l), HGB increased (11.3 instead of 8.9 g per dl) and HCT, MCH and MCHC all improved to nearly normal values, despite the persistence of occult bleeding. Thus, one injection of peptides derived from natural casein seemed capable of stimulating erythropoiesis and reducing anemia associated with blood loss in humans.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Obesity (AREA)
  • Zoology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des peptides actifs biologiquement dérivés de séquences identiques au N-terminus de la fraction αS1 de la caséine du lait, ou similaires à ces séquences. Ces peptides sont en mesure de stimuler et de renforcer la réaction immunitaire, de protéger contre des infections virales, de normaliser les taux de cholestérol sérique, et de stimuler l'hématopoïèse. Les peptides dérivés de la caséine ne sont pas toxiques et peuvent être utilisés dans le traitement et dans la prévention de pathologies immunitaires, de l'hypercholestérolémie, de troubles hématologiques et de maladies virales.
EP02758768A 2001-08-30 2002-08-29 Peptides derives de la caseine et leurs utilisations therapeutiques Withdrawn EP1556074A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US942121 2001-08-30
US09/942,121 US20020147144A1 (en) 2000-03-01 2001-08-30 Casein derived peptides and uses thereof in therapy
PCT/IL2002/000720 WO2003018606A2 (fr) 2001-08-30 2002-08-29 Peptides derives de la caseine et leurs utilisations therapeutiques

Publications (2)

Publication Number Publication Date
EP1556074A2 true EP1556074A2 (fr) 2005-07-27
EP1556074A4 EP1556074A4 (fr) 2008-05-07

Family

ID=25477603

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02758768A Withdrawn EP1556074A4 (fr) 2001-08-30 2002-08-29 Peptides derives de la caseine et leurs utilisations therapeutiques

Country Status (15)

Country Link
EP (1) EP1556074A4 (fr)
JP (1) JP2005511499A (fr)
KR (1) KR20040078639A (fr)
CN (1) CN1694719A (fr)
AU (1) AU2002324323A2 (fr)
BR (1) BR0212625A (fr)
CA (1) CA2458924A1 (fr)
CZ (1) CZ2004335A3 (fr)
HU (1) HUP0500995A3 (fr)
IL (1) IL160548A0 (fr)
MX (1) MXPA04001890A (fr)
NO (1) NO20040880L (fr)
PL (1) PL375113A1 (fr)
WO (1) WO2003018606A2 (fr)
ZA (1) ZA200401574B (fr)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0313892D0 (en) * 2003-06-16 2003-07-23 Hannah Res Inst Control of lactation
AU2005209845A1 (en) * 2004-01-30 2005-08-18 Chemgenex Pharmaceuticals, Inc. Naphthalimide dosing by N-acetyl transferase genotyping
CN101124261A (zh) * 2004-03-01 2008-02-13 派普特拉医药有限公司 酪蛋白衍生肽及其治疗用途
GB0423352D0 (en) 2004-10-21 2004-11-24 Hannah Res Inst "Control of mammary cell number"
ES2319475B1 (es) * 2005-06-08 2010-02-16 Consejo Superior Investig. Cientificas Peptidos bioactivos identificados en hidrolizados enzimaticos de caseinas lacteas y procedimiento de obtencion.
CN101410131B (zh) * 2006-06-09 2012-09-12 森永乳业株式会社 脂质代谢改善剂
JP2011026213A (ja) * 2009-07-22 2011-02-10 Snow Brand Milk Prod Co Ltd 1型糖尿病抑制ペプチド
EP2839291B1 (fr) * 2012-04-16 2018-07-04 The Cleveland Clinic Foundation Vaccin multivalent contre le cancer du sein
CN110381964A (zh) * 2017-03-03 2019-10-25 森永乳业株式会社 Glp-1分泌促进剂和组合物
CN107814835B (zh) * 2017-12-01 2020-04-28 熊猫乳品集团股份有限公司 一种生物活性多肽avpitptlnreq及其制备方法和应用
CN108017709B (zh) * 2017-12-12 2020-04-10 浙江辉肽生命健康科技有限公司 一种生物活性多肽kepmigvnqela及其制备方法和应用
CN113952446B (zh) * 2021-09-03 2023-12-05 河南省医药科学研究院 生物活性肽在抑制骨髓毒性中的应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2118134A1 (fr) * 1970-12-16 1972-07-28 Assad Najjar V
WO2001064234A1 (fr) * 2000-03-01 2001-09-07 Chay 13 Medical Research Group N.V. Peptides derivant de la caseine et leurs emplois therapeutiques

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6150547A (en) * 1997-03-21 2000-11-21 Snow Brand Milk Products Co., Ltd. Iron-casein complex hydrolyzate and process for the production thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2118134A1 (fr) * 1970-12-16 1972-07-28 Assad Najjar V
WO2001064234A1 (fr) * 2000-03-01 2001-09-07 Chay 13 Medical Research Group N.V. Peptides derivant de la caseine et leurs emplois therapeutiques

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KARIYA K ET AL: "INHIBITION OF ANGIOTENSIN CONVERTING ENZYME OF RAT BRAIN WITH BRADYKININ AND ITS FRAGMENTS" BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 100, no. 1, 1981, pages 31-36, XP008066034 ISSN: 0006-291X *
See also references of WO03018606A2 *

Also Published As

Publication number Publication date
WO2003018606A9 (fr) 2004-04-08
WO2003018606A2 (fr) 2003-03-06
WO2003018606A3 (fr) 2005-05-06
CN1694719A (zh) 2005-11-09
EP1556074A4 (fr) 2008-05-07
HUP0500995A2 (en) 2007-05-02
ZA200401574B (en) 2007-02-28
CA2458924A1 (fr) 2003-03-06
MXPA04001890A (es) 2004-06-18
AU2002324323A2 (en) 2003-03-10
JP2005511499A (ja) 2005-04-28
IL160548A0 (en) 2004-07-25
NO20040880L (no) 2004-04-20
HUP0500995A3 (en) 2008-09-29
KR20040078639A (ko) 2004-09-10
BR0212625A (pt) 2007-06-19
CZ2004335A3 (cs) 2004-07-14
PL375113A1 (en) 2005-11-28

Similar Documents

Publication Publication Date Title
AU782662B2 (en) Casein derived peptides and uses thereof in therapy
EP1751179A2 (fr) Peptides derives de la caseine et leurs utilisations therapeutiques
US7666996B2 (en) Casein derived peptides and uses thereof
WO2003018606A2 (fr) Peptides derives de la caseine et leurs utilisations therapeutiques
US7396681B1 (en) Application of Hsp70 proteins
JPH10509422A (ja) 幹細胞増殖インヒビターおよびその使用
JPH10502371A (ja) リンホトキシンを用いる癌処置法
JP2004508412A (ja) 抗新生物薬および免疫刺激薬としてのチオニン

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040313

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

A4 Supplementary search report drawn up and despatched

Effective date: 20080403

17Q First examination report despatched

Effective date: 20080714

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090127