US20080306009A1 - Immunoregulatory compositions - Google Patents

Immunoregulatory compositions Download PDF

Info

Publication number
US20080306009A1
US20080306009A1 US12/069,401 US6940108A US2008306009A1 US 20080306009 A1 US20080306009 A1 US 20080306009A1 US 6940108 A US6940108 A US 6940108A US 2008306009 A1 US2008306009 A1 US 2008306009A1
Authority
US
United States
Prior art keywords
seq
nmpf
amino acid
acid
peptides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/069,401
Inventor
Nisar Ahmed Khan
Robbert Benner
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.)
Erasmus Universiteit Rotterdam
Biotempt BV
Original Assignee
Biotempt BV
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 PCT/NL2001/000259 external-priority patent/WO2001072831A2/en
Priority claimed from US10/028,075 external-priority patent/US20030113733A1/en
Application filed by Biotempt BV filed Critical Biotempt BV
Priority to US12/069,401 priority Critical patent/US20080306009A1/en
Assigned to ERASMUS UNIVERSITEIT ROTTERDAM reassignment ERASMUS UNIVERSITEIT ROTTERDAM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BENNER, ROBBERT, KHAN, NISAR AHMED
Assigned to BIOTEMPT B.V. reassignment BIOTEMPT B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROTTERDAM, ERASMUS UNIVERSITEIT
Publication of US20080306009A1 publication Critical patent/US20080306009A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • 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
    • 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
    • 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
    • C07K14/575Hormones
    • C07K14/59Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K4/00Peptides having up to 20 amino acids in an undefined or only partially defined sequence; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0806Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/081Tripeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1021Tetrapeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1024Tetrapeptides with the first amino acid being heterocyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates generally to biotechnology, and, more specifically, to compositions having immunoregulatory activity, which compounds include particular oligopeptides derived from human chorionic gonadotropin (hCG).
  • hCG human chorionic gonadotropin
  • 6,620,416, U.S. Pat. No. 6,596,688, WO 01/11048 A2, WO 01/10907 A2, and U.S. Pat. No. 6,583,109 relate to various oligopeptides and their use in, among other things, “inhibiting HIV infection,” “treating or preventing HIV infection,” “treating or preventing cancer,” “treating or preventing a condition characterized by loss of body cell mass,” “treating or preventing a condition associated with pathological angiogenesis,” “treating or preventing hematopoietic deficiency,” “ex vivo gene therapy,” “expanding blood cells in vitro,” and/or “providing blood cells to a subject.”
  • compositions containing purified or isolated oligopeptides described herein have immunoregulatory activity useful in, for example, the treatment of sepsis and other disease states and conditions. They also have gene regulatory activities.
  • compositions comprising a purified or isolated peptide consisting of particular four to eight amino acid segments of the sequence MTRVLQGVLPALPQVVC (SEQ ID NO:44 of the incorporated herein SEQUENCE LISTING); and derivatives thereof having one or more conservative substitutions relative to the sequence of SEQ ID NO:44.
  • the particular compositions exhibit immunoregulatory activity as determined by measuring the segment's ability to modulate production of NO by a cell.
  • the compositions have the ability to decrease shock in a subject (e.g., a mammal) undergoing sepsis.
  • the amino acid segment includes a tetrameric sequence (corresponding to the LQVG (SEQ ID NO:1) portion of SEQ ID NO:44, i.e.,
  • AAL AAQ AAG AAV wherein AAL is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Ala and Leu; AAQ is a substituted or unsubstituted amino acid selected from the group consisting of Gln, Pro, and Ala; AAG is a substituted or unsubstituted Gly; and AAV is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Val and Ala.
  • the peptide could be selected from the group consisting of LQGV (SEQ ID NO:1), the derivative AQGV (SEQ ID NO:2), the derivative LQGA (SEQ ID NO:19), the derivative LAGV (SEQ ID NO:26), and the derivative LPGC (SEQ ID NO:41).
  • the segment is the tetramer MTRV (SEQ ID NO:42) or QVVC (SEQ ID NO:43).
  • the segment is six or seven amino acids long, and comprises the sequence
  • AAV AAL Pro Arg AAL2 AAP wherein AAV is substituted or unsubstituted Val or Ala, wherein AAL and AAL2 are independently selected from substituted or unsubstituted Lys or Ala, and wherein AAP is a substituted or unsubstituted Pro or Ala.
  • the purified or isolated peptide can have a formula selected from the group consisting of VLPALP (SEQ ID NO:3), the derivative ALPALP (SEQ ID NO:21), the derivative VAPALP (SEQ ID NO:22), the derivative ALPALPQ (SEQ ID NO:23), the derivative VLPAAPQ (SEQ ID NO:24), the derivative VLPALAQ (SEQ ID NO:25), the derivative VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), the derivative VLPALPA (SEQ ID NO:31), the derivative GVLPALP (SEQ ID NO:32), and the derivative VLAALP (SEQ ID NO:117).
  • VLPALP SEQ ID NO:3
  • the derivative ALPALP SEQ ID NO:21
  • the derivative VAPALP SEQ ID NO:22
  • the derivative ALPALPQ SEQ ID NO:23
  • the derivative VLPAAPQ SEQ ID NO:24
  • the derivative VLPALAQ SEQ ID NO:25
  • the composition has no more than eight amino acids, and includes an amino acid sequence consisting of:
  • AAL AAQ AAG AAV wherein AAL is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Ala, Leu, and Met, wherein AAQ is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Gln, Thr, Ala, and Pro, wherein AAG is substituted or unsubstituted Gly or Arg, and wherein AAV is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Cys, Ala, and Val.
  • AAL is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Ala, Leu, and Met
  • AAQ is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Gln, Thr, Ala, and Pro
  • AAG is substituted or unsubstituted Gly or Arg
  • AAV is a substituted or unsubstitute
  • the sequence is preferably selected from the group consisting of Leu Gln Gly Val (SEQ ID NO:1), Ala Gln Gly Val (SEQ ID NO:2), Leu Gln Gly Ala (SEQ ID NO:19), Leu Ala Gly Val (SEQ ID NO:26), Leu Pro Gly Cys (SEQ ID NO:41), and Met Thr Arg Val (SEQ ID NO:42).
  • the composition is the sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:19, SEQ ID NO:26, SEQ ID NO:41, SEQ ID NO:42, or a salt of any thereof.
  • composition comprising a purified or isolated peptide, or acid addition salt thereof, the purified or isolated peptide (a) consisting of an amino acid sequence selected from the group consisting of: (i) a four to seven amino acid segment of the sequence of MTRVLQGVLPALPQVVC (SEQ ID NO:44); and (ii) a derivative of the segment of (a) having one or more conservative substitutions relative to the sequence of SEQ ID NO:44; and (b) exhibiting an immunoregulatory activity as determined by measuring its capability of modulating production of NO by a cell.
  • a purified or isolated peptide consisting of an amino acid sequence selected from the group consisting of: (i) a four to seven amino acid segment of the sequence of MTRVLQGVLPALPQVVC (SEQ ID NO:44); and (ii) a derivative of the segment of (a) having one or more conservative substitutions relative to the sequence of SEQ ID NO:44; and (b) exhibiting an immunoregulatory activity as determined by measuring
  • Such a pharmaceutical composition preferably includes a sequence selected from the group selected from the group consisting of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), VLPALP (SEQ ID NO:3), LQGA (SEQ ID NO:19), ALPALP (SEQ ID NO:21), VAPALP (SEQ ID NO:22), ALPALPQ (SEQ ID NO:23), VLPAAPQ (SEQ ID NO:24), VLPALAQ (SEQ ID NO:25), LAGV (SEQ ID NO:26), VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), VLPALPA (SEQ ID NO:31), and GVLPALP (SEQ ID NO:32), LPGC (SEQ ID NO:41), MTRV (SEQ ID NO:42), QVVC (SEQ ID NO:43), VLAALP (SEQ ID NO:117), and combinations of any thereof, with or without other active or inactive ingredients, presented in a pharmaceutically acceptable
  • the invention is a purified or isolated peptide consisting of GVLPALPQ (SEQ ID NO:33), or an acid addition salt thereof.
  • the invention would thus also include a pharmaceutical composition comprising the peptide of SEQ ID NO:33 or an addition salt thereof, together with a pharmaceutically acceptable excipient.
  • the invention comprises a purified or isolated peptide (or, for example, acid addition salt thereof), selected from the group consisting of AQG, MTR, and VVC.
  • the invention also includes a composition comprising one or more of the following amino acid segments: LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), VLPALP (SEQ ID NO:3), LQGA (SEQ ID NO:19), ALPALP (SEQ ID NO:21), VAPALP (SEQ ID NO:22), ALPALPQ (SEQ ID NO:23), VLPAAPQ (SEQ ID NO:24), VLPALAQ (SEQ ID NO:25), LAGV (SEQ ID NO:26), VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), VLPALPA (SEQ ID NO:31), and GVLPALP (SEQ ID NO:32), LPGC (SEQ ID NO:41), MTRV (SEQ ID NO:42), QVVC (SEQ ID NO:43), VLAALP (SEQ ID NO:117), AQG, MTR, or VVC.
  • LQGV SEQ ID NO:1
  • included is a composition comprising a purified or isolated peptide consisting of sequence LQG in an amount sufficient to exhibit an immunoregulatory activity as determined by measuring the sequence LQG's ability to modulate production of NO by a cell.
  • a method of treating bone disease comprising administering to a subject believed to be in need of such treatment a composition comprising an oligopeptide, derivative or functional analogue thereof, the particular molecule capable of modulating production of NO and/or TNF-alpha by a cell.
  • Such a method of treatment is particularly useful in post-menopausal women that no longer experience the benefits of being provided with a natural source of hCG and its breakdown products.
  • Such a treatment can be achieved by systemic administration of a composition of the invention according to the invention, but local administration in joints, bursa or tendon sheaths is provided as well.
  • the molecule can be selected from Table 6 or identified in a method described herein.
  • the treatment comprises administering to the subject a pharmaceutical composition comprising an oligopeptide or functional analogue thereof capable of reducing production of NO by a cell, for example, wherein the composition comprises at least two oligopeptides or functional analogues thereof, each capable of reducing production of NO and/or TNF-alpha by a cell, in particular wherein the at least two oligopeptides are selected from the group LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), and VLPALP (SEQ ID NO:3).
  • LQGV SEQ ID NO:1
  • AQGV SEQ ID NO:2
  • VLPALP VLPALP
  • oligopeptides hereof have been tested, both ex vivo and in vivo, and in small animals.
  • Immunomodulatory effects with these oligopeptides have been observed in vitro and in ex vivo such as in T-cell assays showing the inhibition of pathological Th1 immune responses, suppression of inflammatory cytokines (MIF), increase in production of anti-inflammatory cytokines (IL-10, TGF-beta) and immunomodulatory effects on antigen-presenting cells (APC) like dendritic cells, monocytes and macrophages.
  • MIF inflammatory cytokines
  • APC antigen-presenting cells
  • oligopeptides as provided herein allows for rational design of signal molecule mixtures that better alleviate the symptoms seen with sepsis.
  • One such mixture a 1:1:1 mixture of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2) and VLPALP (SEQ ID NO:3) was administered to primates in a gram-negative-induced rhesus monkey sepsis model for prevention of septic shock and found to be effective in this primate model.
  • the invention provides a pharmaceutical composition for the treatment of sepsis in a primate and a method for the treatment of sepsis in a primate comprising subjecting the primate to a composition of the invention according to the invention, preferably to a mixture of such composition of the inventions.
  • Administration of such a composition of the invention or mixture preferably occurs systematically, for example, by intravenous or intraperitoneal administration.
  • such treatment also comprises the use of for example an antibiotic, however, only when such use is not contra indicated because of the risk of generating further toxin loads because of lysis of the bacteria subject to the action of those antibiotics in an individual thus treated.
  • compositions according to the invention for the preparation of a pharmaceutical composition or medicament and methods of treating various medical conditions that are other than use in the preparation of a pharmaceutical composition for the treatment of an immune-mediated disorder or a method of treatment of an immune-mediated disorder or treatment of a wasting syndrome.
  • a “purified or isolated” peptide is one that has been purified from a natural or biotechnological source, or, more preferably, is synthesized as described herein.
  • composition refers to chemical compounds which contain or consist of the oligopeptide.
  • the oligopeptide is preferably isolated before inclusion within the composition.
  • the oligopeptide most preferably consists of three (3) to six (6) amino acids.
  • the previously described preferred compound could, in one embodiment be:
  • NT at the N-terminus is selected from the group of H—, CH 3 —, an acyl group, or a general protective group; and CT at the C-terminus is selected from the group of small (e.g., one to five amino acids) peptides, —OH, —OR1, —NH2, —NHR1, —NR1R2, or —N(CH2)1-6 NR1 R2, wherein R1 and R2, when present, are independently selected from H, alkyl, aryl, alkyl, aralkyl, and wherein R1 and R2 can be cyclically bonded to one another.
  • small (e.g., one to five amino acids) peptides —OH, —OR1, —NH2, —NHR1, —NR1R2, or —N(CH2)1-6 NR1 R2, wherein R1 and R2, when present, are independently selected from H, alkyl, aryl, alkyl, aralkyl, and wherein R1 and R2 can
  • Alkyl is preferably a saturated branched or unbranched hydrocarbon having one to six carbon atoms, e.g., methyl, ethyl, and isopentyl.
  • Aryl is an aromatic hydrocarbon group, preferably having six to ten carbon atoms, such as phenyl or naphthyl.
  • (Ar)alkyl is an arene group (having both aliphatic and aromatic portions), preferably having seven to 13 carbon atoms such as benzyl, ethylbenzyl, n-propylbenzyl, and isobutylbenzyl.
  • Oletypeptide as used herein are peptides having from three to eight amino acids joined together by peptide bonds. Equivalent to oligopeptides are compounds having the same or equivalent side chains as the particular amino acids used in an oligopeptide, and arranged sequentially in the same order as the peptides, but joined together by non-peptide bonds, e.g., by isosteric linkages such as the keto isostere, hydroxy isostere, diketo isostere, or the keto-difluoromethylene isostere.
  • composition also includes, for example, an acceptable salt of the oligopeptide or a labeled oligopeptide.
  • acceptable salt refers to salts that retain the desired activity of the oligopeptide or equivalent compound, but preferably do not detrimentally affect the activity of the oligopeptide or other component of a system in which uses the oligopeptide.
  • examples of such salts are acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfinuric acid, phosphoric acid, nitric acid, and the like.
  • Salts may also be formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, and the like.
  • Salts may be formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel and the like or with an organic cation formed from N,N′-dibenzylethylenediamine or ethylenediamine, or combinations thereof (e.g., a zinc tannate salt).
  • the thus developed chemical entity can be administered and introduced in-vivo systemically, topically, or locally.
  • the peptide, or is modification or derivative can be administered as the entity as such or as a pharmaceutically acceptable acid- or base addition salt, formed by reaction with an inorganic acid (such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid); or with an organic acid (such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid); or by reaction with an inorganic base (such as sodium hydroxide, ammonium hydroxide, potassium hydroxide); or with an organic base (such as mono-, di-, trialkyl and aryl amines and substituted ethanolamines).
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, perch
  • a selected peptide and any of the derived entities may also be conjugated to sugars, lipids, other polypeptides, nucleic acids and PNA; and function in-situ as a conjugate or be released locally after reaching a targeted tissue or organ.
  • substitutions with regard to the various amino acids generally relate to substituting a group such as alkoxy, halogen, hydroxy, nitro, or lower alkyl onto an aromatic ring for a hydrogen that would usually be present. Substitutions can also be made on the alkyl chain connecting the aromatic portion to the peptide backbone, with, for instance lower alkyl groups substituting for a hydrogen. Still further substitutions can be made at the alpha position of an amino acid, also using an alkyl group.
  • Substitutions with regard to the amino acid phenylalanine include compounds such as L/D-homophenylalanyl, N methyl phenylalanyl, .alpha.-methylphenylalanyl, and .alpha.-methyl-tyrosyl.
  • substitutions involve the use of fluorine or chlorine as a halogen, and methoxy as an alkoxy group.
  • alkyl and lower alkyl generally alkyl groups having fewer (one to three) carbon atoms are preferred.
  • the compounds according to the general formula may be prepared in a manner conventional for such compounds.
  • suitably N alpha. protected (and side-chain protected if reactive side-chains are present) amino acid derivatives or peptides are activated and coupled to suitably carboxyl protected amino acid or peptide derivatives either in solution or on a solid support. Protection of the .alpha.-amino functions generally takes place by urethane functions such as the acid-labile tertiary-butyloxycarbonyl group (“Boc”), benzyloxycarbonyl (“Z”) group and substituted analogs or the base-labile 9-fluoremyl-methyloxycarbonyl (“Fmoc”) group.
  • Boc acid-labile tertiary-butyloxycarbonyl group
  • Z benzyloxycarbonyl
  • Fmoc base-labile 9-fluoremyl-methyloxycarbonyl
  • the Z group can also be removed by catalytic hydrogenation,
  • suitable protecting groups include the Nps, Bmv, Bpoc, Aloc, MSC, etc.
  • a good overview of amino protecting groups is given in The peptides, Analysis, Synthesis, Biology , Vol. 3 E. Gross and J. Meienhofer, eds. (Academic Press, New York, 1981). Protection of carboxyl groups can take place by ester formation, for example, base-labile esters like methyl or ethyl, acid labile esters like tert. butyl or, substituted, benzyl esters or hydrogenolytically. Protection of side-chain functions like those of lysine and glutamic or aspartic acid can take place using the aforementioned groups.
  • Activation of the carboxyl group of the suitably protected amino acids or peptides can take place by the azide, mixed anhydride, active ester, or carbodiimide method especially with the addition of catalytic and racemization-suppressing compounds like 1-N—N-hydroxybenzotriazole, N-hydroxysuccinimide, 3-hydroxy-4-oxo-3,4-dihydro-1,2,3,-benzotriazine, N-hydroxy-5 norbornene-2,3-dicar-boxyimide.
  • the anhydrides of phosphorus based acids can be used. See, e.g., The Peptides, Analysis, Synthesis, Biology , supra and Pure and Applied Chemistry, 59(3), 331-344 (1987).
  • Removal of the protecting groups, and, in the case of solid phase peptide synthesis, the cleavage from the solid support, can take place in different ways, depending on the nature of those protecting groups and the type of linker to the solid support. Usually deprotection takes place under acidic conditions and in the presence of scavengers. See, e.g., Volumes 3, 5 and 9 of the series on The Peptides Analysis, Synthesis, Biology , supra.
  • oligopeptides according to the invention could also be made according to recombinant DNA methods. Such methods involve the preparation of the desired oligopeptide thereof by means of expressing recombinant polynucleotide sequence which codes for one or more of the oligopeptides in question in a suitable microorganism as host.
  • the process involves introducing into a cloning vehicle (e.g., a plasmid, phage DNA, or other DNA sequence able to replicate in a host cell) a DNA sequence coding for the particular oligopeptide or oligopeptides, introducing the cloning vehicle into a suitable eucaryotic or prokaryotic host cell, and culturing the host cell thus transformed.
  • a cloning vehicle e.g., a plasmid, phage DNA, or other DNA sequence able to replicate in a host cell
  • the cloning vehicle e.g., a plasmid, phage DNA, or other DNA sequence able to replicate in a host cell
  • the cloning vehicle e.g., a plasmid, phage DNA, or other DNA sequence able to replicate in a host cell
  • the cloning vehicle e.g., a plasmid, phage DNA, or other DNA sequence able to
  • a “functional analogue” or “derivative” of a peptide includes an amino acid sequence, or other sequence monomers, which has been altered such that the functional properties of the sequence are essentially the same in kind, not necessarily in amount.
  • An analogue or derivative can be provided in many ways, for instance, through “conservative amino acid substitution.”
  • peptidomimetic compounds can be designed that functionally or structurally resemble the original peptide taken as the starting point but that are for example composed of non-naturally occurring amino acids or polyamides. With “conservative amino acid substitution,” one amino acid residue is substituted with another residue with generally similar properties (size, hydrophobicity), such that the overall functioning is likely not to be seriously affected.
  • a derivative can also be provided by systematically improving at least one desired property of an amino acid sequence. This can, for instance, be done by an Ala-scan and/or replacement net mapping method. With these methods, many different peptides are generated, based on an original amino acid sequence but each containing a substitution of at least one amino acid residue. The amino acid residue may either be replaced by alanine (Ala-scan) or by any other amino acid residue (replacement net mapping). This way, many positional variants of the original amino acid sequence are synthesized. Every positional variant is screened for a specific activity. The generated data are used to design improved peptide derivatives of a certain amino acid sequence.
  • a derivative or analogue can also be, for instance, generated by substitution of an L-amino acid residue with a D-amino acid residue.
  • This substitution leading to a peptide which does not naturally occur in nature, can improve a property of an amino acid sequence. It is, for example, useful to provide a peptide sequence of known activity of all D-amino acids in retro inversion format, thereby allowing for retained activity and increased half-life values.
  • improved peptide derivatives comprising such D-amino acids can be designed with further improved characteristics.
  • peptides or analogues can be circularized, for example, by providing them with (terminal) cysteines, dimerized or multimerized, for example, by linkage to lysine or cysteine or other compounds with side-chains that allow linkage or multimerization, brought in tandem- or repeat-configuration, conjugated or otherwise linked to carriers known in the art, if only by a labile link that allows dissociation.
  • Synthetic versions of these oligopeptides as described above, and functional analogues or derivatives of these breakdown products, are herein provided to modulate gene expression in a cell and be used in methods to rectify errors in gene expression or the treatment of disease.
  • composition as used herein is intended to cover both the active composition of the invention alone or a composition containing the composition of the invention together with a pharmaceutically acceptable carrier, diluent or excipient.
  • Acceptable diluents of an oligopeptide as described herein in the detailed description are for example physiological salt solutions or phosphate buffered salt solutions.
  • a signal molecule is administered in an effective concentration to an animal or human systemically, e.g., by intravenous, intramuscular or intraperitoneal administration.
  • Another way of administration comprises perfusion of organs or tissue, be it in vivo or ex vivo, with a perfusion fluid comprising a signal molecule according to the invention.
  • Topical administration may also apply, e.g., in inflammations of the skin or mucosal surfaces of for example mouth, nose and/or genitals.
  • Local administration can occur in joints, bursa, tendon sheaths, in or around the spinal cord at locations where nerve bundles branch off, at the location of hernias, in or around infarcted areas in brain or heart, etc.
  • the administration may be done as a single dose, as a discontinuous sequence of various doses, or continuously for a period of time sufficient to permit substantial modulation of gene expression. In the case of a continuous administration, the duration of the administration may vary depending upon a number of factors which would readily be appreciated by those skilled in the art.
  • the administration dose of the active molecule may be varied over a fairly broad range.
  • concentrations of an active molecule which can be administered would be limited by efficacy at the lower end and the solubility of the compound at the upper end.
  • the optimal dose or doses for a particular patient should and can be determined by the physician or medical specialist involved, taking into consideration well-known relevant factors such as the condition, weight and age of the patient, etc.
  • the active molecule may be administered directly in a suitable vehicle, such as, for example, phosphate-buffered saline (PBS) or solutions in alcohol or DMSO.
  • a suitable vehicle such as, for example, phosphate-buffered saline (PBS) or solutions in alcohol or DMSO.
  • PBS phosphate-buffered saline
  • the active molecule is administered through a single dose delivery using a drug-delivery system, such as a sustained-release delivery system, which enables the maintenance of the required concentrations of the active molecule for a period of time sufficient for adequate modulation of gene expression.
  • a suitable drug-delivery system would be pharmacologically inactive or at least tolerable. It should preferably not be immunogenic nor cause inflammatory reactions, and should permit release of the active molecule so as to maintain effective levels thereof over the desired time period.
  • Suitable delivery vehicles include, but are not limited to, the following: microcapsules or microspheres; liposomes and other lipid-based release systems; viscous instillates; absorbable and/or biodegradable mechanical barriers and implants; and polymeric delivery materials, such as polyethylene oxide/polypropylene oxide block copolymers, polyesters, cross-linked polyvinylalcohols, polyanhydrides, polymethacrylate and polymethacrylamide hydrogels, anionic carbohydrate polymers, etc.
  • Useful delivery systems are well known in the art.
  • a highly suitable formulation to achieve the active molecule release comprises injectable microcapsules or microspheres made from a biodegradable polymer, such as poly(dl-lactide), poly(dl-lactide-co-glycolide), polycaprolactone, polyglycolide, polylactic acid-co-glycolide, poly(hydroxybutyric acid), polyesters or polyacetals.
  • injectable systems comprising microcapsules or microspheres having a diameter of about 50 to about 500 micrometers offer advantages over other delivery systems. For example, they generally use less active molecules and may be administered by paramedical personnel. Moreover, such systems are inherently flexible in the design of the duration and rate of separate drug release by selection of microcapsule or microsphere size, drug loading and dosage administered. Further, they can be successfully sterilized by gamma irradiation.
  • microcapsules and microspheres are well within the reach of persons skilled in the art and detailed information concerning these points is available in the literature.
  • Biodegradable polymers such as lactide, glycolide and caprolactone polymers
  • Fibers or filaments comprising the active molecule are also contemplated as within the scope of the present invention.
  • Another highly suitable formulation for a single-dose delivery of the active molecule in accordance with the present invention involves liposomes.
  • the encapsulation of an active molecule in liposomes or multilamellar vesicles is a well-known technique for targeted drug delivery and prolonged drug residence.
  • the preparation and use of drug-loaded liposomes is well within the reach of persons skilled in the art and well documented in the literature.
  • Suitable approach for single-dose delivery of an active molecule in accordance with the present invention involves the use of viscous instillates.
  • high molecular weight carriers are used in admixture with the active molecule, giving rise to a structure which produces a solution with high viscosity.
  • Suitable high molecular weight carriers include, but are not limited to, the following: dextrans and cyclodextrans; hydrogels; (cross-linked) viscous materials, including (cross-linked) viscoelastics; carboxymethylcellulose; hyaluronic acid; and chondroitin sulfate.
  • the active molecule may be administered in combination with absorbable mechanical barriers such as oxidized regenerated cellulose.
  • the active molecule may be covalently or non-covalently (e.g., ionically) bound to such a barrier, or it may simply be dispersed therein.
  • the side-chain of glutamine was protected with a trityl function.
  • the peptides were synthesized manually. Each coupling consisted of the following steps: (i) removal of the alpha-amino Fmoc-protection by piperidine in dimethylformamide (DMF), (ii) coupling of the Fmoc amino acid (3 eq) with diisopropylcarbodiimide (DIC)/1-hydroxybenzotriazole (HOBt) in DMF/N-methylformamide (NMP) and (iii) capping of the remaining amino functions with acetic anhydride/diisopropylethylamine (DIEA) in DMF/NMP.
  • DMF dimethylformamide
  • DIEA acetic anhydride/diisopropylethylamine
  • the peptide resin was treated with a mixture of trifluoroacetic acid (TFA)/H 2 O/triisopropylsilane (TIS) 95:2.5:2.5. After 30 minutes, TIS was added until decolorization. The solution was evaporated in vacuo and the peptide precipitated with diethylether.
  • TFA trifluoroacetic acid
  • TIS triisopropylsilane
  • VLPALP SEQ ID NO:3
  • VLPALPQ SEQ ID NO:29
  • the collected fractions were concentrated to about 5 ml by rotation film evaporation under reduced pressure at 40° C.
  • the remaining TFA was exchanged against acetate by eluting two times over a column with anion exchange resin (Merck II) in acetate form.
  • the eluate was concentrated and lyophilized in 28 hours. Peptides later were prepared for use by dissolving them in PBS.
  • mice 255957 from NV Organon of Oss, NL) and with synthetic peptides (5 mg/kg) after two hours of LPS injection.
  • BALB/c mice were injected i.p. either with 10 mg/kg or with 11 mg/kg LPS ( E. coli 026:B6; Difco Lab., Detroit, Mich., USA). Subsequently, mice were treated after two hours and 24 hours of LPS treatment with NMPF peptides.
  • Derivatives of these peptides for example (but not limited to), by addition of classical and non-classical amino acids or derivatives that are differentially modified during or after synthesis, for example benzylation, amidation, glycosylation, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc., could also lead to a better effectiveness of the activity.
  • NMPF synthetic peptides
  • mice treated with PBS after the shock induction reached a sickness score of 5 at 14 and 24 hours, and remained so after the second injection with PBS.
  • the survival rate in control group mice was 0% at 48 hours.
  • mice treated with NMPF 9, 11, 12, 43, 46, 50 and 60 reached a maximum sickness score of 2-3 at 24 hours after the induction of septic shock and further reached a maximum sickness score of 1-2 at 48 hours after the second injection of NMPF.
  • mice treated with NMPF 5, 7, 8, 45, 53 and 58 reached a sickness score of 5 and after the second injection with NMPF all mice returned to a sickness score of 1-2 and survival rates in NMPF groups were 100%.
  • NMPF peptides have anti-shock activity at different stages of the disease and NMPF have anti-shock activity even at the disease stage when otherwise irreversible damage had been done. This indicates that NMPF have effects on different cellular levels and also have repairing and regenerating capacity.
  • mice Every four days, urine was checked for the presence of glucose (Gluketur Test; Boehringer Mannheim, Mannheim, Del.). All mice used in these studies were maintained in a pathogen-free facility. They were given free access to food and water. The experiments were approved by the Animal Experiments Committee of the Erasmus University Rotterdam. Diabetes was assessed by measurement of the venous blood glucose level using an Abbott Medisense Precision glucometer. Mice were considered diabetic after two consecutive glucose measurements ⁇ 11 mmol/l (200 mg/dl). Onset of diabetes was dated from the first consecutive reading.
  • the RAW 264.7 murine macrophage cell line obtained from American Type Culture Collection (Manassas, Va., USA), were cultured at 37° C. in 5% CO 2 using DMEM containing 10% fetal calf serum (FCS), 50 U/ml penicillin, 50 ⁇ g/ml streptomycin, 0.2 M Na-pyruvate, 2 mM glutamine and 50 ⁇ M 2-mercaptoethanol (Bio Whittaker, Europe). The medium was changed every two days.
  • FCS fetal calf serum
  • penicillin 50 ⁇ g/ml streptomycin
  • 0.2 M Na-pyruvate 2 mM glutamine
  • 2-mercaptoethanol Bio Whittaker, Europe
  • Nitrite measurements Nitrite production was measured in the RAW 264.7 macrophage supernatants.
  • the cells (7.5 ⁇ 10 5 /ml) were cultured in 48-well plates in 500 ⁇ l of culture medium. The cells were stimulated with LPS (10 microg/ml) and/or NMPF (1 pg/ml, 1 ng/ml, 1 ⁇ g/ml) for 24 hours, then the culture media were collected.
  • Nitrite was measured by adding 100 microl of Griess reagent (Sigma) to 100 microl samples of culture medium. The OD 540 was measured using a microplate reader, and the nitrite concentration was calculated by comparison with the OD 540 produced using standard solutions of sodium nitrite in the culture medium.
  • NMPF neuropeptide
  • NOD mice treated three times a week for two weeks with NMPF showed profound inhibition of diabetes development.
  • the strongest anti-diabetic effects were seen with NMPF-1, -4, -5, -6, -7, -65, -66 and commercial hCG preparation (PREGNYL®, batch no. 235863).
  • NMPF-71 showed no effect on the incidence of diabetes, while NMPF-64 and NMPF-11 had a moderate anti-diabetic effect.
  • NO production is a central mediator of the vascular and inflammatory response.
  • macrophages RAW 264.7
  • LPS LPS-induced NMPF peptides
  • these cells co-stimulated with most of the NMPF peptides (NMPF peptides 1 to 14, 43 to 66 and 69) even in a very low dose (1 pg/ml) inhibited the production of NO.
  • the invention provides a method and a composition for the treatment of conditions that are associated with dysfunctional LDL receptors such as apoE and other members of the apolipoprotein family.
  • a composition of the invention comprising GVLPALPQ (SEQ ID NO:33) (NMPF-5) and/or VLPALP (SEQ ID NO:3) (NMPF-6) or a functional analogue or derivative thereof is preferred.
  • VLPALPQVVC 100 50 17 (SEQ ID NO:20) 2 LQGVLPALPQ 100 67 0 0 (SEQ ID NO:49) 3 LQG 100 83 20 17 4 LQGV 100 100 100 100 (SEQ ID NO:1) 5 GVLPALPQ 100 100 80 17 (SEQ ID NO:33) 6 VLPALP 100 100 100 100 (SEQ ID NO:3) 7 VLPALPQ 100 83 0 0 (SEQ ID NO:168) 8 GVLPALP 100 100 83 67 (SEQ ID NO:32) 9 VVC 100 100 50 50 11 MTRV 100 100 67 50 (SEQ ID NO:42) 12 MTR 100 100 100 67 50 13 LQGVLPALPQVVC 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100
  • NMPF a mixture 1:1:1 of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2) and VLPALP (SEQ ID NO:3), administered in a gram-negative-induced rhesus monkey sepsis model for prevention of septic shock.
  • Two rhesus monkeys were infused with a 10 10 CFU per kg of the gram-negative bacterium E. coli to induce a fatal septic shock.
  • One monkey received placebo-treatment and was sacrificed within seven hours after infusion of the bacteria without recovery from the anesthesia.
  • the second monkey received treatment with test compound and was sacrificed at the same time point.
  • the monkeys were kept anesthetized throughout the observation period and sacrificed seven hours after the start of the bacterium infusion for pathological examination.
  • the animals underwent a gross necropsy in which the abdominal and thorax cavities were opened and internal organs examined in situ.
  • the Escherichia coli strain was purchased from ATCC ( E. coli; 086a: K61 serotype, ATCC 33985). In a control experiment, the strain proved equally susceptible to bactericidal factors in human and rhesus monkey serum. Prior to the experiment, a fresh culture was set-up; the E. coli strain was cultured for one day, harvested and washed thoroughly to remove free endotoxin. Prior to infusion into the animal, the number and viability of the bacteria were assessed. Serial dilutions of the E. coli stock were plated on BHI agar and cultured overnight at 37° C. The colonies on each plate were counted and the number of colony-forming units per ml was calculated.
  • the body weight measurement of the day of the experiment was used to calculate the E. coli dose and E. coli stock was suspended in isotonic saline (N.P.B.I., Emmer-Compascuum, NL) at the concentration needed for infusion (total dose volume for infusion approximately 10 ml/kg.
  • the E. coli suspension was kept on ice until infusion.
  • Antibiotic was used to synchronize the shock induction in the monkeys.
  • Baytril (Baytril 2.5%, Bayer, Del.) was used instead of gentamycin, as the strain proved only marginally susceptible to the latter antibiotic.
  • Individual animals were identified by a number or letter combination tattooed on the chest.
  • the femoral or the cephalic vein was cannulated and used for infusing isotonic saline, live E. coli and antibiotic administration. Insensible fluid loss was compensated for by infusing isotonic saline containing 2.5% glucose (Fresenius, 's Hertogenbosch, NL) at a rate of 3.3 ml/kg/hour.
  • Preparative actions During anesthesia the animals were instrumented for measurement of blood pressure (with an automatic cuff), heart rate and body temperature. Isotonic saline was infused at 3.3 ml/kg/hour to compensate for fluid loss. Femoral vessels were cannulated for infusion of E. coli and antibiotics. Temperature-controlled heating pads were used to support body temperature. The monkeys were continuously monitored during the E. coli challenge and for the six-hour period following E. coli administration. After seven hours, two animals (the control animal and one treated with NMPF) were sacrificed to compare the direct effect of the compound at the level of histology. The third animal, treated with NMPF, was allowed to recover from anesthesia and was intensively observed during the first 12 hours after recovery followed by frequent daily observation. The decision to allow the third animal to recover was made after consulting with the veterinarian.
  • Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg).
  • NMPF NMPF oligopeptides
  • LQGV LQGV
  • AQGV SEQ ID NO:2
  • VLPALP SEQ ID NO:3
  • kidneys The following organs were weighed and a bacterial count was performed: kidneys, liver, lungs, lymph nodes, and gross lesions.
  • Tissues of all organs were preserved in neutral aqueous phosphate buffered 4% solution of formaldehyde. Lymphoid organs were cryopreserved. All tissues will be processed for histopathological examination.
  • this bacterial dose induced lethal shock within eight hours after the start of the infusion.
  • the infusion period was two hours.
  • Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg).
  • the monkey was observed by the authorized veterinarian without knowing which of the monkeys received NMPF treatment.
  • NMPF Monkey 459
  • E. coli 086 10 CFU/kg
  • this bacterial dose induced lethal shock within eight hours after the start of the infusion.
  • the infusion period was two hours.
  • NMPF was i.v. injected in a single bolus injection.
  • Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg). After the E. coli injection, this monkey was also observed by the authorized veterinarian without knowing which of the monkeys received NMPF treatment.

Abstract

The invention relates to compounds exhibiting immunoregulatory activity as determined by measuring the compound's ability to modulate production of NO by a cell. Preferred compounds include or consist of a sequence
AAL AAQ AAG AAV

wherein AAL is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Ala and Leu; wherein AAQ is a substituted or unsubstituted amino acid selected from the group consisting of Gln, Pro, and Ala; wherein AAG is a substituted or unsubstituted amino acid Gly, and wherein AAV is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Val and Ala. In certain embodiments, the compound consists of a tripeptide selected from the group AQG, MTR, VVC, and mixtures thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of co-pending application Ser. No. 10/753,510, filed Jan. 7, 2004, U.S. patent Ser. No. ______, which is a continuation in part of U.S. Ser. No. 10/028,075, filed Dec. 21, 2001, a continuation of U.S. Ser. No. 10/262,522, filed Sep. 30, 2002, U.S. patent Ser. No. ______, which is a continuation of International Application No. PCT/NL01/00259, designating the United States of America published, in English, as PCT International Publication No. WO 01/72831 A2 filed Mar. 3, 2001; and application Ser. No. 10/753,510 is further a continuation in part of International Application No. PCT/NL02/00639, designating the United States of America and published on Apr. 10, 2003, in English, as PCT International Publication No. WO 03/029292 A2. The contents of each of the herein referenced U.S. patent applications and PCT patent publications are incorporated herein by this reference in their entirety.
  • TECHNICAL FIELD
  • The invention relates generally to biotechnology, and, more specifically, to compositions having immunoregulatory activity, which compounds include particular oligopeptides derived from human chorionic gonadotropin (hCG).
  • BACKGROUND
  • U.S. Pat. No. 5,380,668 to Herron (Jan. 10, 1995), the contents of the entirety of which are incorporated by this reference, discloses, among other things, various compounds having the antigenic binding activity of hCG. The oligopeptides disclosed therein are disclosed generally for use in diagnostic methods.
  • Various patents and patent applications to Gallo et al. (e.g., U.S. Pat. No. 5,677,275 (corresponding to WO 96/04008 A1), U.S. Pat. No. 5,877,148 (also corresponding to WO 96/04008 A1), WO 97/49721 A1, U.S. Pat. No. 6,319,504 (corresponding to WO 97/49373), U.S. Patent Application 2003/0049273 A1 (also corresponding to WO 97/49373), U.S. Pat. No. 5,968,513 (corresponding to WO 97/49418), U.S. Pat. No. 5,997,871 (corresponding to WO 97/49432), U.S. Pat. No. 6,620,416, U.S. Pat. No. 6,596,688, WO 01/11048 A2, WO 01/10907 A2, and U.S. Pat. No. 6,583,109) relate to various oligopeptides and their use in, among other things, “inhibiting HIV infection,” “treating or preventing HIV infection,” “treating or preventing cancer,” “treating or preventing a condition characterized by loss of body cell mass,” “treating or preventing a condition associated with pathological angiogenesis,” “treating or preventing hematopoietic deficiency,” “ex vivo gene therapy,” “expanding blood cells in vitro,” and/or “providing blood cells to a subject.”
  • DISCLOSURE OF THE INVENTION
  • As we described in PCT International Publication No. WO 03/029292 A2 (published Apr. 10, 2003), PCT International Publication No. WO 01/72831 A2 (published Oct. 4, 2001), and U.S. Pat. No. 6,844,315 (issued Jan. 18, 2005), U.S. Pat. No. 7,175,679 (issued Feb. 13, 2007), and U.S. Patent Application 20030113733 A1 (published Jun. 19, 2003), and 20030166556 A1 (published Sep. 4, 2003), the contents of all of which are incorporated by this reference, compositions containing purified or isolated oligopeptides described herein have immunoregulatory activity useful in, for example, the treatment of sepsis and other disease states and conditions. They also have gene regulatory activities.
  • Provided is a composition comprising a purified or isolated peptide consisting of particular four to eight amino acid segments of the sequence MTRVLQGVLPALPQVVC (SEQ ID NO:44 of the incorporated herein SEQUENCE LISTING); and derivatives thereof having one or more conservative substitutions relative to the sequence of SEQ ID NO:44. The particular compositions exhibit immunoregulatory activity as determined by measuring the segment's ability to modulate production of NO by a cell. Preferably, the compositions have the ability to decrease shock in a subject (e.g., a mammal) undergoing sepsis.
  • In certain embodiments, the amino acid segment includes a tetrameric sequence (corresponding to the LQVG (SEQ ID NO:1) portion of SEQ ID NO:44, i.e.,
  • AAL AAQ AAG AAV

    wherein AAL is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Ala and Leu; AAQ is a substituted or unsubstituted amino acid selected from the group consisting of Gln, Pro, and Ala; AAG is a substituted or unsubstituted Gly; and AAV is a substituted or unsubstituted non-polar amino acid selected from the group consisting of Val and Ala. For instance, the peptide could be selected from the group consisting of LQGV (SEQ ID NO:1), the derivative AQGV (SEQ ID NO:2), the derivative LQGA (SEQ ID NO:19), the derivative LAGV (SEQ ID NO:26), and the derivative LPGC (SEQ ID NO:41).
  • In a different embodiment, the segment is the tetramer MTRV (SEQ ID NO:42) or QVVC (SEQ ID NO:43).
  • In certain embodiments, the segment is six or seven amino acids long, and comprises the sequence
  • AAV AAL Pro Arg AAL2 AAP

    wherein AAV is substituted or unsubstituted Val or Ala, wherein AAL and AAL2 are independently selected from substituted or unsubstituted Lys or Ala, and wherein AAP is a substituted or unsubstituted Pro or Ala.
  • In such an embodiment, the purified or isolated peptide can have a formula selected from the group consisting of VLPALP (SEQ ID NO:3), the derivative ALPALP (SEQ ID NO:21), the derivative VAPALP (SEQ ID NO:22), the derivative ALPALPQ (SEQ ID NO:23), the derivative VLPAAPQ (SEQ ID NO:24), the derivative VLPALAQ (SEQ ID NO:25), the derivative VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), the derivative VLPALPA (SEQ ID NO:31), the derivative GVLPALP (SEQ ID NO:32), and the derivative VLAALP (SEQ ID NO:117).
  • In certain embodiments, the composition has no more than eight amino acids, and includes an amino acid sequence consisting of:
  • AAL AAQ AAG AAV

    wherein AAL is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Ala, Leu, and Met, wherein AAQ is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Gln, Thr, Ala, and Pro, wherein AAG is substituted or unsubstituted Gly or Arg, and wherein AAV is a substituted or unsubstituted amino acid selected from the group of amino acids consisting of Cys, Ala, and Val. Again such a composition is characterized in having immunoregulatory activity as determined by measuring its capability of modulating production of NO by a cell.
  • In such an embodiment, the sequence is preferably selected from the group consisting of Leu Gln Gly Val (SEQ ID NO:1), Ala Gln Gly Val (SEQ ID NO:2), Leu Gln Gly Ala (SEQ ID NO:19), Leu Ala Gly Val (SEQ ID NO:26), Leu Pro Gly Cys (SEQ ID NO:41), and Met Thr Arg Val (SEQ ID NO:42). Preferably, the composition is the sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:19, SEQ ID NO:26, SEQ ID NO:41, SEQ ID NO:42, or a salt of any thereof.
  • Further included is a pharmaceutical composition comprising a purified or isolated peptide, or acid addition salt thereof, the purified or isolated peptide (a) consisting of an amino acid sequence selected from the group consisting of: (i) a four to seven amino acid segment of the sequence of MTRVLQGVLPALPQVVC (SEQ ID NO:44); and (ii) a derivative of the segment of (a) having one or more conservative substitutions relative to the sequence of SEQ ID NO:44; and (b) exhibiting an immunoregulatory activity as determined by measuring its capability of modulating production of NO by a cell.
  • Such a pharmaceutical composition preferably includes a sequence selected from the group selected from the group consisting of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), VLPALP (SEQ ID NO:3), LQGA (SEQ ID NO:19), ALPALP (SEQ ID NO:21), VAPALP (SEQ ID NO:22), ALPALPQ (SEQ ID NO:23), VLPAAPQ (SEQ ID NO:24), VLPALAQ (SEQ ID NO:25), LAGV (SEQ ID NO:26), VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), VLPALPA (SEQ ID NO:31), and GVLPALP (SEQ ID NO:32), LPGC (SEQ ID NO:41), MTRV (SEQ ID NO:42), QVVC (SEQ ID NO:43), VLAALP (SEQ ID NO:117), and combinations of any thereof, with or without other active or inactive ingredients, presented in a pharmaceutically acceptable form for administration to a human.
  • In one preferred embodiment, the invention is a purified or isolated peptide consisting of GVLPALPQ (SEQ ID NO:33), or an acid addition salt thereof. The invention would thus also include a pharmaceutical composition comprising the peptide of SEQ ID NO:33 or an addition salt thereof, together with a pharmaceutically acceptable excipient.
  • In another embodiment the invention comprises a purified or isolated peptide (or, for example, acid addition salt thereof), selected from the group consisting of AQG, MTR, and VVC.
  • The invention also includes a composition comprising one or more of the following amino acid segments: LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), VLPALP (SEQ ID NO:3), LQGA (SEQ ID NO:19), ALPALP (SEQ ID NO:21), VAPALP (SEQ ID NO:22), ALPALPQ (SEQ ID NO:23), VLPAAPQ (SEQ ID NO:24), VLPALAQ (SEQ ID NO:25), LAGV (SEQ ID NO:26), VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), VLPALPA (SEQ ID NO:31), and GVLPALP (SEQ ID NO:32), LPGC (SEQ ID NO:41), MTRV (SEQ ID NO:42), QVVC (SEQ ID NO:43), VLAALP (SEQ ID NO:117), AQG, MTR, or VVC.
  • In certain embodiments, included is a composition comprising a purified or isolated peptide consisting of sequence LQG in an amount sufficient to exhibit an immunoregulatory activity as determined by measuring the sequence LQG's ability to modulate production of NO by a cell.
  • Provided is a method of treating bone disease, such as osteoporosis, the method comprising administering to a subject believed to be in need of such treatment a composition comprising an oligopeptide, derivative or functional analogue thereof, the particular molecule capable of modulating production of NO and/or TNF-alpha by a cell.
  • Such a method of treatment is particularly useful in post-menopausal women that no longer experience the benefits of being provided with a natural source of hCG and its breakdown products. Such a treatment can be achieved by systemic administration of a composition of the invention according to the invention, but local administration in joints, bursa or tendon sheaths is provided as well. The molecule can be selected from Table 6 or identified in a method described herein. The treatment comprises administering to the subject a pharmaceutical composition comprising an oligopeptide or functional analogue thereof capable of reducing production of NO by a cell, for example, wherein the composition comprises at least two oligopeptides or functional analogues thereof, each capable of reducing production of NO and/or TNF-alpha by a cell, in particular wherein the at least two oligopeptides are selected from the group LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), and VLPALP (SEQ ID NO:3).
  • Several oligopeptides hereof have been tested, both ex vivo and in vivo, and in small animals. A beneficial effect of these oligopeptides on LPS-induced sepsis in mice, namely the inhibition of the effect of the sepsis, was observed. Immunomodulatory effects with these oligopeptides have been observed in vitro and in ex vivo such as in T-cell assays showing the inhibition of pathological Th1 immune responses, suppression of inflammatory cytokines (MIF), increase in production of anti-inflammatory cytokines (IL-10, TGF-beta) and immunomodulatory effects on antigen-presenting cells (APC) like dendritic cells, monocytes and macrophages.
  • Now knowing the gene modulatory effect of the composition of the inventions such as oligopeptides as provided herein allows for rational design of signal molecule mixtures that better alleviate the symptoms seen with sepsis. One such mixture, a 1:1:1 mixture of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2) and VLPALP (SEQ ID NO:3) was administered to primates in a gram-negative-induced rhesus monkey sepsis model for prevention of septic shock and found to be effective in this primate model. Accordingly, the invention provides a pharmaceutical composition for the treatment of sepsis in a primate and a method for the treatment of sepsis in a primate comprising subjecting the primate to a composition of the invention according to the invention, preferably to a mixture of such composition of the inventions. Administration of such a composition of the invention or mixture preferably occurs systematically, for example, by intravenous or intraperitoneal administration. In certain embodiments, such treatment also comprises the use of for example an antibiotic, however, only when such use is not contra indicated because of the risk of generating further toxin loads because of lysis of the bacteria subject to the action of those antibiotics in an individual thus treated.
  • Also provided is the use of a composition according to the invention for the preparation of a pharmaceutical composition or medicament and methods of treating various medical conditions that are other than use in the preparation of a pharmaceutical composition for the treatment of an immune-mediated disorder or a method of treatment of an immune-mediated disorder or treatment of a wasting syndrome.
  • DETAILED DESCRIPTION OF THE INVENTION
  • As used herein, a “purified or isolated” peptide is one that has been purified from a natural or biotechnological source, or, more preferably, is synthesized as described herein.
  • “Composition,” as used herein, refers to chemical compounds which contain or consist of the oligopeptide. The oligopeptide is preferably isolated before inclusion within the composition. The oligopeptide most preferably consists of three (3) to six (6) amino acids.
  • For instance, the previously described preferred compound could, in one embodiment be:
  • NT AA1 AA2 AA3 AA4 CT

    wherein NT at the N-terminus is selected from the group of H—, CH3—, an acyl group, or a general protective group; and CT at the C-terminus is selected from the group of small (e.g., one to five amino acids) peptides, —OH, —OR1, —NH2, —NHR1, —NR1R2, or —N(CH2)1-6 NR1 R2, wherein R1 and R2, when present, are independently selected from H, alkyl, aryl, alkyl, aralkyl, and wherein R1 and R2 can be cyclically bonded to one another.
  • “Alkyl,” as used herein, is preferably a saturated branched or unbranched hydrocarbon having one to six carbon atoms, e.g., methyl, ethyl, and isopentyl.
  • “Aryl,” as used herein, is an aromatic hydrocarbon group, preferably having six to ten carbon atoms, such as phenyl or naphthyl.
  • “(Ar)alkyl,” as used herein, is an arene group (having both aliphatic and aromatic portions), preferably having seven to 13 carbon atoms such as benzyl, ethylbenzyl, n-propylbenzyl, and isobutylbenzyl.
  • “Oligopeptide,” as used herein are peptides having from three to eight amino acids joined together by peptide bonds. Equivalent to oligopeptides are compounds having the same or equivalent side chains as the particular amino acids used in an oligopeptide, and arranged sequentially in the same order as the peptides, but joined together by non-peptide bonds, e.g., by isosteric linkages such as the keto isostere, hydroxy isostere, diketo isostere, or the keto-difluoromethylene isostere.
  • “Composition” also includes, for example, an acceptable salt of the oligopeptide or a labeled oligopeptide. As used herein, “acceptable salt” refers to salts that retain the desired activity of the oligopeptide or equivalent compound, but preferably do not detrimentally affect the activity of the oligopeptide or other component of a system in which uses the oligopeptide. Examples of such salts are acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfinuric acid, phosphoric acid, nitric acid, and the like. Salts may also be formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, and the like. Salts may be formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel and the like or with an organic cation formed from N,N′-dibenzylethylenediamine or ethylenediamine, or combinations thereof (e.g., a zinc tannate salt).
  • The thus developed chemical entity can be administered and introduced in-vivo systemically, topically, or locally. The peptide, or is modification or derivative, can be administered as the entity as such or as a pharmaceutically acceptable acid- or base addition salt, formed by reaction with an inorganic acid (such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid); or with an organic acid (such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid); or by reaction with an inorganic base (such as sodium hydroxide, ammonium hydroxide, potassium hydroxide); or with an organic base (such as mono-, di-, trialkyl and aryl amines and substituted ethanolamines). A selected peptide and any of the derived entities may also be conjugated to sugars, lipids, other polypeptides, nucleic acids and PNA; and function in-situ as a conjugate or be released locally after reaching a targeted tissue or organ.
  • A “substitution” with regard to the various amino acids generally relate to substituting a group such as alkoxy, halogen, hydroxy, nitro, or lower alkyl onto an aromatic ring for a hydrogen that would usually be present. Substitutions can also be made on the alkyl chain connecting the aromatic portion to the peptide backbone, with, for instance lower alkyl groups substituting for a hydrogen. Still further substitutions can be made at the alpha position of an amino acid, also using an alkyl group.
  • Substitutions with regard to the amino acid phenylalanine include compounds such as L/D-homophenylalanyl, N methyl phenylalanyl, .alpha.-methylphenylalanyl, and .alpha.-methyl-tyrosyl.
  • Preferred substitutions involve the use of fluorine or chlorine as a halogen, and methoxy as an alkoxy group. With regard to alkyl and lower alkyl, generally alkyl groups having fewer (one to three) carbon atoms are preferred.
  • The compounds according to the general formula may be prepared in a manner conventional for such compounds. To that end, suitably N alpha. protected (and side-chain protected if reactive side-chains are present) amino acid derivatives or peptides are activated and coupled to suitably carboxyl protected amino acid or peptide derivatives either in solution or on a solid support. Protection of the .alpha.-amino functions generally takes place by urethane functions such as the acid-labile tertiary-butyloxycarbonyl group (“Boc”), benzyloxycarbonyl (“Z”) group and substituted analogs or the base-labile 9-fluoremyl-methyloxycarbonyl (“Fmoc”) group. The Z group can also be removed by catalytic hydrogenation, Other suitable protecting groups include the Nps, Bmv, Bpoc, Aloc, MSC, etc. A good overview of amino protecting groups is given in The peptides, Analysis, Synthesis, Biology, Vol. 3 E. Gross and J. Meienhofer, eds. (Academic Press, New York, 1981). Protection of carboxyl groups can take place by ester formation, for example, base-labile esters like methyl or ethyl, acid labile esters like tert. butyl or, substituted, benzyl esters or hydrogenolytically. Protection of side-chain functions like those of lysine and glutamic or aspartic acid can take place using the aforementioned groups. Protection of thiol, and although not always required, of guanidino, alcohol and imidazole groups can take place using a variety of reagents such as those described in The Peptides, Analysis, Synthesis, Biology, id. or in Pure and Applied Chemistry, 59(3), 331-344 (1987). Activation of the carboxyl group of the suitably protected amino acids or peptides can take place by the azide, mixed anhydride, active ester, or carbodiimide method especially with the addition of catalytic and racemization-suppressing compounds like 1-N—N-hydroxybenzotriazole, N-hydroxysuccinimide, 3-hydroxy-4-oxo-3,4-dihydro-1,2,3,-benzotriazine, N-hydroxy-5 norbornene-2,3-dicar-boxyimide. Also the anhydrides of phosphorus based acids can be used. See, e.g., The Peptides, Analysis, Synthesis, Biology, supra and Pure and Applied Chemistry, 59(3), 331-344 (1987).
  • It is also possible to prepare the compounds by the solid phase method of Merrifield. Different solid supports and different strategies are known see, e.g., Barany and Merrifield in The Peptides, Analysis, Synthesis, Biology, Vol. 2, E. Gross and J. Meienhofer, eds. (Acad. Press, New York, 1980), Kneib-Cordonier and Mullen, Int. J. Peptide Protein Res., 30, 705-739 (1987), and Fields and Noble, Int. J. Peptide Protein Res., 35, 161-214 (1990). The synthesis of compounds in which a peptide bond is replaced by an isostere, can, in general, be performed using the previously described protecting groups and activation procedures. Procedures to synthesize the modified isosteres are described in the literature, e.g., for the —CH2—NH— isostere and for the —CO—CH2— isostere.
  • Removal of the protecting groups, and, in the case of solid phase peptide synthesis, the cleavage from the solid support, can take place in different ways, depending on the nature of those protecting groups and the type of linker to the solid support. Usually deprotection takes place under acidic conditions and in the presence of scavengers. See, e.g., Volumes 3, 5 and 9 of the series on The Peptides Analysis, Synthesis, Biology, supra.
  • Another possibility is the application of enzymes in synthesis of such compounds; for reviews see, e.g., H. D. Jakubke in The Peptides, Analysis, Synthesis, Biology, Vol. 9, S. Udenfriend and J. Meienhofer, eds. (Acad. Press, New York, 1987).
  • Although possibly not desirable from an economic point of view, oligopeptides according to the invention could also be made according to recombinant DNA methods. Such methods involve the preparation of the desired oligopeptide thereof by means of expressing recombinant polynucleotide sequence which codes for one or more of the oligopeptides in question in a suitable microorganism as host. Generally, the process involves introducing into a cloning vehicle (e.g., a plasmid, phage DNA, or other DNA sequence able to replicate in a host cell) a DNA sequence coding for the particular oligopeptide or oligopeptides, introducing the cloning vehicle into a suitable eucaryotic or prokaryotic host cell, and culturing the host cell thus transformed. In certain embodiments, the compound may include a glycoprotein portion.
  • As used herein, a “functional analogue” or “derivative” of a peptide includes an amino acid sequence, or other sequence monomers, which has been altered such that the functional properties of the sequence are essentially the same in kind, not necessarily in amount. An analogue or derivative can be provided in many ways, for instance, through “conservative amino acid substitution.” Also peptidomimetic compounds can be designed that functionally or structurally resemble the original peptide taken as the starting point but that are for example composed of non-naturally occurring amino acids or polyamides. With “conservative amino acid substitution,” one amino acid residue is substituted with another residue with generally similar properties (size, hydrophobicity), such that the overall functioning is likely not to be seriously affected. However, it is often much more desirable to improve a specific function. A derivative can also be provided by systematically improving at least one desired property of an amino acid sequence. This can, for instance, be done by an Ala-scan and/or replacement net mapping method. With these methods, many different peptides are generated, based on an original amino acid sequence but each containing a substitution of at least one amino acid residue. The amino acid residue may either be replaced by alanine (Ala-scan) or by any other amino acid residue (replacement net mapping). This way, many positional variants of the original amino acid sequence are synthesized. Every positional variant is screened for a specific activity. The generated data are used to design improved peptide derivatives of a certain amino acid sequence.
  • A derivative or analogue can also be, for instance, generated by substitution of an L-amino acid residue with a D-amino acid residue. This substitution, leading to a peptide which does not naturally occur in nature, can improve a property of an amino acid sequence. It is, for example, useful to provide a peptide sequence of known activity of all D-amino acids in retro inversion format, thereby allowing for retained activity and increased half-life values. By generating many positional variants of an original amino acid sequence and screening for a specific activity, improved peptide derivatives comprising such D-amino acids can be designed with further improved characteristics.
  • A person skilled in the art is well able to generate analogous compounds of an amino acid sequence. This can, for instance, be done through screening of a peptide library. Such an analogue has essentially the same functional properties of the sequence in kind, not necessarily in amount. Also, peptides or analogues can be circularized, for example, by providing them with (terminal) cysteines, dimerized or multimerized, for example, by linkage to lysine or cysteine or other compounds with side-chains that allow linkage or multimerization, brought in tandem- or repeat-configuration, conjugated or otherwise linked to carriers known in the art, if only by a labile link that allows dissociation.
  • Synthetic versions of these oligopeptides as described above, and functional analogues or derivatives of these breakdown products, are herein provided to modulate gene expression in a cell and be used in methods to rectify errors in gene expression or the treatment of disease.
  • The term “pharmaceutical composition” as used herein is intended to cover both the active composition of the invention alone or a composition containing the composition of the invention together with a pharmaceutically acceptable carrier, diluent or excipient. Acceptable diluents of an oligopeptide as described herein in the detailed description are for example physiological salt solutions or phosphate buffered salt solutions. In certain embodiments, a signal molecule is administered in an effective concentration to an animal or human systemically, e.g., by intravenous, intramuscular or intraperitoneal administration. Another way of administration comprises perfusion of organs or tissue, be it in vivo or ex vivo, with a perfusion fluid comprising a signal molecule according to the invention. Topical administration, e.g., in ointments or sprays, may also apply, e.g., in inflammations of the skin or mucosal surfaces of for example mouth, nose and/or genitals. Local administration can occur in joints, bursa, tendon sheaths, in or around the spinal cord at locations where nerve bundles branch off, at the location of hernias, in or around infarcted areas in brain or heart, etc. The administration may be done as a single dose, as a discontinuous sequence of various doses, or continuously for a period of time sufficient to permit substantial modulation of gene expression. In the case of a continuous administration, the duration of the administration may vary depending upon a number of factors which would readily be appreciated by those skilled in the art.
  • The administration dose of the active molecule may be varied over a fairly broad range. The concentrations of an active molecule which can be administered would be limited by efficacy at the lower end and the solubility of the compound at the upper end. The optimal dose or doses for a particular patient should and can be determined by the physician or medical specialist involved, taking into consideration well-known relevant factors such as the condition, weight and age of the patient, etc.
  • The active molecule may be administered directly in a suitable vehicle, such as, for example, phosphate-buffered saline (PBS) or solutions in alcohol or DMSO. Pursuant to preferred embodiments of the present invention, however, the active molecule is administered through a single dose delivery using a drug-delivery system, such as a sustained-release delivery system, which enables the maintenance of the required concentrations of the active molecule for a period of time sufficient for adequate modulation of gene expression. A suitable drug-delivery system would be pharmacologically inactive or at least tolerable. It should preferably not be immunogenic nor cause inflammatory reactions, and should permit release of the active molecule so as to maintain effective levels thereof over the desired time period. Alternatives are known in the art as suitable for purposes of sustained release and are contemplated as within the scope of the present invention. Suitable delivery vehicles include, but are not limited to, the following: microcapsules or microspheres; liposomes and other lipid-based release systems; viscous instillates; absorbable and/or biodegradable mechanical barriers and implants; and polymeric delivery materials, such as polyethylene oxide/polypropylene oxide block copolymers, polyesters, cross-linked polyvinylalcohols, polyanhydrides, polymethacrylate and polymethacrylamide hydrogels, anionic carbohydrate polymers, etc. Useful delivery systems are well known in the art.
  • A highly suitable formulation to achieve the active molecule release comprises injectable microcapsules or microspheres made from a biodegradable polymer, such as poly(dl-lactide), poly(dl-lactide-co-glycolide), polycaprolactone, polyglycolide, polylactic acid-co-glycolide, poly(hydroxybutyric acid), polyesters or polyacetals. Injectable systems comprising microcapsules or microspheres having a diameter of about 50 to about 500 micrometers offer advantages over other delivery systems. For example, they generally use less active molecules and may be administered by paramedical personnel. Moreover, such systems are inherently flexible in the design of the duration and rate of separate drug release by selection of microcapsule or microsphere size, drug loading and dosage administered. Further, they can be successfully sterilized by gamma irradiation.
  • The design, preparation, and use of microcapsules and microspheres are well within the reach of persons skilled in the art and detailed information concerning these points is available in the literature. Biodegradable polymers (such as lactide, glycolide and caprolactone polymers) may also be used in formulations other than microcapsules and microspheres; e.g., premade films and spray-on films of these polymers containing the active molecule would be suitable for use in accordance with the present invention. Fibers or filaments comprising the active molecule are also contemplated as within the scope of the present invention.
  • Another highly suitable formulation for a single-dose delivery of the active molecule in accordance with the present invention involves liposomes. The encapsulation of an active molecule in liposomes or multilamellar vesicles is a well-known technique for targeted drug delivery and prolonged drug residence. The preparation and use of drug-loaded liposomes is well within the reach of persons skilled in the art and well documented in the literature.
  • Yet another suitable approach for single-dose delivery of an active molecule in accordance with the present invention involves the use of viscous instillates. In this technique, high molecular weight carriers are used in admixture with the active molecule, giving rise to a structure which produces a solution with high viscosity. Suitable high molecular weight carriers include, but are not limited to, the following: dextrans and cyclodextrans; hydrogels; (cross-linked) viscous materials, including (cross-linked) viscoelastics; carboxymethylcellulose; hyaluronic acid; and chondroitin sulfate. The preparation and use of drug-loaded viscous instillates is well known to persons skilled in the art.
  • Pursuant to yet another approach, the active molecule may be administered in combination with absorbable mechanical barriers such as oxidized regenerated cellulose. The active molecule may be covalently or non-covalently (e.g., ionically) bound to such a barrier, or it may simply be dispersed therein.
  • The invention is further explained with the aid of the following illustrative examples.
  • EXAMPLES Example I Material and Methods
  • Peptide Synthesis: The peptides as mentioned herein such as LQG, AQG, LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2), LQGA (SEQ ID NO:19), VLPALP (SEQ ID NO:13), ALPALP (SEQ ID NO:21), VAPALP (SEQ ID NO:22), ALPALPQ (SEQ ID NO:23), VLPAAPQ (SEQ ID NO:24), VLPALAQ (SEQ ID NO:25), LAGV (SEQ ID NO:26), VLAALP (SEQ ID NO:27), VLPALA (SEQ ID NO:28), VLPALPQ (SEQ ID NO:29), VLAALPQ (SEQ ID NO:30), VLPALPA (SEQ ID NO:31), GVLPALP (SEQ ID NO:32), VVCNYRDVRFESIRLPGCPRGVNPVVSYAVALSCQCAL (SEQ ID NO:35), RPRCRPINATLAVEKEGCPVCITVNTTICAGYCPT (SEQ ID NO:45), SKAPPPSLPSPSRLPGPS (SEQ ID NO:38), LQGVLPALPQVVC (SEQ ID NO:34), SIRLPGCPRGVNPVVS (SEQ ID NO:39), LPGCPRGVNPVVS (SEQ ID NO:40), LPGC (SEQ ID NO:41), MTRV (SEQ ID NO:42), MTR, and VVC were prepared by solid-phase synthesis (R. B. Merrifield, J. Am. Chem. Soc., 85:2149-2165 (1963)) using the fluorenylmethoxycarbonyl (Fmoc)/tert-butyl-based methodology (Atherton, 1985) with 2-chlorotrityl chloride resin (Barlos et al., Int. J. Peptide Protein Res., 37:513-520 (1991)) as the solid support.
  • The side-chain of glutamine was protected with a trityl function. The peptides were synthesized manually. Each coupling consisted of the following steps: (i) removal of the alpha-amino Fmoc-protection by piperidine in dimethylformamide (DMF), (ii) coupling of the Fmoc amino acid (3 eq) with diisopropylcarbodiimide (DIC)/1-hydroxybenzotriazole (HOBt) in DMF/N-methylformamide (NMP) and (iii) capping of the remaining amino functions with acetic anhydride/diisopropylethylamine (DIEA) in DMF/NMP. Upon completion of the synthesis, the peptide resin was treated with a mixture of trifluoroacetic acid (TFA)/H2O/triisopropylsilane (TIS) 95:2.5:2.5. After 30 minutes, TIS was added until decolorization. The solution was evaporated in vacuo and the peptide precipitated with diethylether.
  • The crude peptides were dissolved in water (50-100 mg/ml) and purified by reverse-phase high-performance liquid chromatography (RP-HPLC). HPLC conditions were: column: Vydac TP21810C18 (10×250 mm); elution system: gradient system of 0.1% TFA in water v/v (A) and 0.1% TFA in acetonitrile (ACN) v/v (B); flow rate 6 ml/minute; absorbance was detected from 190-370 nm. There were different gradient systems used. For example, for peptides LQG and LQGV (SEQ ID NO:1): ten minutes 100% A followed by linear gradient 0-10% B in 50 minutes. For example, for peptides VLPALP (SEQ ID NO:3) and VLPALPQ (SEQ ID NO:29): five minutes 5% B followed by linear gradient 1% B/minute. The collected fractions were concentrated to about 5 ml by rotation film evaporation under reduced pressure at 40° C. The remaining TFA was exchanged against acetate by eluting two times over a column with anion exchange resin (Merck II) in acetate form. The eluate was concentrated and lyophilized in 28 hours. Peptides later were prepared for use by dissolving them in PBS.
  • Example II Endotoxin Shock Model (Sepsis)
  • Sepsis. For the endotoxin model, BALB/c mice were injected i.p. with 8-9 mg/kg LPS (E. coli 026:B6; Difco Lab., Detroit, Mich., USA). Control groups (PBS) were treated with PBS i.p. only. To test the effect of NMPF from different sources (synthetic, commercial hCG preparation [c-hCG]), we treated BALB/c mice with a dose of 300-700 IU of different hCG preparations (PG23; PREGNYLTM batch no. 235863, PG25; PREGNYLTM batch no. 255957 from NV Organon of Oss, NL) and with synthetic peptides (5 mg/kg) after two hours of LPS injection. In other experiments, BALB/c mice were injected i.p. either with 10 mg/kg or with 11 mg/kg LPS (E. coli 026:B6; Difco Lab., Detroit, Mich., USA). Subsequently, mice were treated after two hours and 24 hours of LPS treatment with NMPF peptides.
  • Semi-quantitative sickness measurements. Mice were scored for sickness severity using the following measurement scheme:
  • 1 Percolated fur, but no detectable behavior differences compared to normal mice.
  • 2 Percolated fur, huddle reflex, responds to stimuli (such as tap on cage), just as active during handling as healthy mouse.
  • Slower response to tap on cage, passive or docile when handled, but still curious when alone in a new setting.
  • 4 Lack of curiosity, little or no response to stimuli, quite immobile.
  • 5 Labored breathing, inability or slow to self-right after being rolled onto back (moribund).
  • 6 Sacrificed.
  • Results Endotoxin Shock Model (Sepsis)
  • Sepsis experiments. To determine the effect of synthetic peptides (NMPF) in high-dose LPS shock model, BALB/c mice were injected intraperitoneally with different doses of LPS and survival was assessed daily for five days. In this experiment (for the LPS endotoxin model), BALB/c mice were injected i.p. with 8-9 mg/kg LPS (E. coli 026:B6; Difco Lab., Detroit, Mich., USA). Control groups (PBS) were treated with PBS i.p. only. We treated BALB/c mice with a dose of 300-700 IU of different hCG preparations (PG23; PREGNYL batch no. 235863, PG25; PREGNYL batch no. 255957) or with peptides (5 mg/kg) after two hours of LPS injection.
  • These experiments showed (Table 1) that NMPF peptides 4, 6, 66 and PG23 inhibited shock completely (all mice had in first 24 hours sickness scores not higher than 2; shortly thereafter they recovered completely and had sickness scores of 0), while peptides 2, 3 and 7 accelerated shock (all mice had in first 24 hours sickness scores of 5 and most of them died, while the control mice treated with LPS+PBS had sickness scores of 3-4 in first 24 hours and most of them died after 48 hours with sickness scores of 5; 17% survival rate at 72 hours). In addition, peptides 1, 5, 8, 9, 11, 12, 13, 14 and 64 showed in a number of different experiments variability in effectiveness as well as in the kind (inhibitory vs accelerating) of activity. This variability is likely attributable to the rate of breakdown of the various peptides and the different effects the various peptides and their breakdown products have in vivo. In addition, these experiments also showed the variability in anti-shock activity in c-hCG preparations that is likely attributable to the variation in the presence of anti-shock and shock-accelerating NMPF. Visible signs of sickness were apparent in all of the experimental animals, but the kinetics and obviously the severity of this sickness were significantly different. These data are representative of at least ten separate experiments.
  • In Table 2, we see the effect of ALA-replacement (PEPSCAN) in peptide LQG, LQGV (SEQ ID NO:1), VLPALP (SEQ ID NO:3), VLPALPQ (SEQ ID NO:29) in septic shock experiments. We conclude that the change in even one amino acid by a neutral amino acid can lead to different activity. So, genomic differences as well as polymorphism in these peptides can regulate the immune response very precisely. Derivatives of these peptides, for example (but not limited to), by addition of classical and non-classical amino acids or derivatives that are differentially modified during or after synthesis, for example benzylation, amidation, glycosylation, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc., could also lead to a better effectiveness of the activity.
  • To determine whether treatment of BALB/c mice with NMPF inhibits septic shock at different stages of disease, synthetic peptides (NMPF) were injected i.p. at two and 24 hours after the induction of septic shock with high dose LPS (10 mg/kg).
  • As shown in Tables 3 and 4, control mice treated with PBS after the shock induction reached a sickness score of 5 at 14 and 24 hours, and remained so after the second injection with PBS. The survival rate in control group mice was 0% at 48 hours. In contrast to control mice, mice treated with NMPF 9, 11, 12, 43, 46, 50 and 60 reached a maximum sickness score of 2-3 at 24 hours after the induction of septic shock and further reached a maximum sickness score of 1-2 at 48 hours after the second injection of NMPF. In addition, mice treated with NMPF 5, 7, 8, 45, 53 and 58 reached a sickness score of 5 and after the second injection with NMPF all mice returned to a sickness score of 1-2 and survival rates in NMPF groups were 100%. Mice treated with NMPF 3 reached sickness scores of 3-4 and the second NMPF injection did save these mice. These experiments show that NMPF peptides have anti-shock activity at different stages of the disease and NMPF have anti-shock activity even at the disease stage when otherwise irreversible damage had been done. This indicates that NMPF have effects on different cellular levels and also have repairing and regenerating capacity.
  • Example III NOD Experiment
  • Mice. Female NOD mice at the age of 13-14 weeks were treated i.p. with PBS (n=6) or NMPF peptides (VLPALPQVVC (SEQ ID NO:20), LQGV (SEQ ID NO:1), GVLPALPQ (SEQ ID NO:33), VLPALP (SEQ ID NO:3), VLPALPQ (SEQ ID NO:29), MTRV (SEQ ID NO:42), LPGCPRGVNPVVS (SEQ ID NO:40), CPRGVNPVVS (SEQ ID NO:50), LPGC (SEQ ID NO:41), MTRVLQGVLPALPQVVC (SEQ ID NO:44), VVCNYRDVRFESIRLPGCPRGVNPVVSYAVALSCQCAL (SEQ ID NO:35)) (5 mg/kg, n=6) three times a week for two weeks. Every four days, urine was checked for the presence of glucose (Gluketur Test; Boehringer Mannheim, Mannheim, Del.). All mice used in these studies were maintained in a pathogen-free facility. They were given free access to food and water. The experiments were approved by the Animal Experiments Committee of the Erasmus University Rotterdam. Diabetes was assessed by measurement of the venous blood glucose level using an Abbott Medisense Precision glucometer. Mice were considered diabetic after two consecutive glucose measurements≦11 mmol/l (200 mg/dl). Onset of diabetes was dated from the first consecutive reading.
  • Glucose tolerance test (GTT) was performed at 28 weeks of age in fasted mice (n=5) by injecting 1 g/kg D-glucose intraperitoneally (i.p.). At 0 (fasting), 5, 30 and 60 minutes, blood samples were collected from the tail and tested for glucose content.
  • Example IV NO Experiment
  • Cell culture. The RAW 264.7 murine macrophage cell line, obtained from American Type Culture Collection (Manassas, Va., USA), were cultured at 37° C. in 5% CO2 using DMEM containing 10% fetal calf serum (FCS), 50 U/ml penicillin, 50 μg/ml streptomycin, 0.2 M Na-pyruvate, 2 mM glutamine and 50 μM 2-mercaptoethanol (Bio Whittaker, Europe). The medium was changed every two days.
  • Nitrite measurements. Nitrite production was measured in the RAW 264.7 macrophage supernatants. The cells (7.5×105/ml) were cultured in 48-well plates in 500 μl of culture medium. The cells were stimulated with LPS (10 microg/ml) and/or NMPF (1 pg/ml, 1 ng/ml, 1 μg/ml) for 24 hours, then the culture media were collected. Nitrite was measured by adding 100 microl of Griess reagent (Sigma) to 100 microl samples of culture medium. The OD540 was measured using a microplate reader, and the nitrite concentration was calculated by comparison with the OD540 produced using standard solutions of sodium nitrite in the culture medium.
  • Results NOD Experiment
  • In order to determine whether NMPF has effect on the disease development in NOD mice, we tested NMPF on pre-diabetic female NOD mice at the age of 13-14 weeks. After only two weeks of treatment (injection of NMPF (5 mg/kg) every other day), glucosuria data of all NOD mice was analyzed at the age of 17 weeks. Profound anti-diabetic effect (mice negative for glucosuria) was observed in different NMPF groups as compared to the PBS group, especially in NMPF groups treated with peptide VLPALPQVVC (SEQ ID NO:20), VLPALP (SEQ ID NO:3), MTRV (SEQ ID NO:42), LPGCPRGVNPVVS (SEQ ID NO:40) and LPGC. In addition, impairment of the glucose tolerance test was positively correlated to insulitis, but negatively correlated to the number of functional beta cells; also this test showed that NOD mice successfully treated with NMPF were tolerant for glucose as compared to the PBS group. Our results show that PBS treated NOD mice were all diabetic at the age of 23 weeks. Whereas, NOD mice treated three times a week for two weeks with NMPF showed profound inhibition of diabetes development. The strongest anti-diabetic effects were seen with NMPF-1, -4, -5, -6, -7, -65, -66 and commercial hCG preparation (PREGNYL®, batch no. 235863). These mice had a low fasting blood glucose level and were tolerant for glucose (data partially shown). However, NMPF-71 showed no effect on the incidence of diabetes, while NMPF-64 and NMPF-11 had a moderate anti-diabetic effect.
  • NO Experiment
  • NO production is a central mediator of the vascular and inflammatory response. Our results show that macrophages (RAW 264.7) stimulated with LPS produce large amounts of NO. However, these cells co-stimulated with most of the NMPF peptides (NMPF peptides 1 to 14, 43 to 66 and 69) even in a very low dose (1 pg/ml) inhibited the production of NO.
  • Results
  • apoE Experiment
  • The invention provides a method and a composition for the treatment of conditions that are associated with dysfunctional LDL receptors such as apoE and other members of the apolipoprotein family. In particular, use of a composition of the invention comprising GVLPALPQ (SEQ ID NO:33) (NMPF-5) and/or VLPALP (SEQ ID NO:3) (NMPF-6) or a functional analogue or derivative thereof is preferred. Groups of apoE-deficient mice (n=6 per group) were fed a high cholesterol food and given PBS or NMPF every other day intraperitoneally. After 2.5 weeks, body weight was determined as shown in the Table below.
  • Average Weight (g) SD (g) p-value
    ApoE−/− PBS 31.667 1.007
    ApoE−/− NMPF-4 31.256 1.496 0.536
    ApoE−/− NMPF-5 29.743 1.160 0.019
    Background/PBS 26.760 1.582 10−6
    ApoE−/− NMPF-6 29.614 1.064 0.004
  • TABLE 1
    Results of shock experiments in mice
    TEST SUBSTANCE % SURVIVAL IN TIME
    (HOURS) 0 16 40 72
    PBS 100 100 67 17
    PG23 100 100 100 100
    PG25 100 83 83 83
    PEPTIDE
    NMPF SEQUENCE
    1 VLPALPQVVC 100 100 50 17
    (SEQ ID NO:20)
    2 LQGVLPALPQ 100 67 0 0
    (SEQ ID NO:49)
    3 LQG 100 83 20 17
    4 LQGV 100 100 100 100
    (SEQ ID NO:1)
    5 GVLPALPQ 100 100 80 17
    (SEQ ID NO:33)
    6 VLPALP 100 100 100 100
    (SEQ ID NO:3)
    7 VLPALPQ 100 83 0 0
    (SEQ ID NO:168)
    8 GVLPALP 100 100 83 67
    (SEQ ID NO:32)
    9 VVC 100 100 50 50
    11 MTRV 100 100 67 50
    (SEQ ID NO:42)
    12 MTR 100 100 67 50
    13 LQGVLPALPQVVC 100 100 100 100
    (SEQ ID NO:34)
    14 (CYCLIC) LQGVLPALPQVVC 100 83 83 83
    (SEQ ID NO:34)
    64 LPGCPRGVNPVVS 100 100 100 100
    (SEQ ID NO:40)
    66 LPGC 100 100 100 100
    (SEQ ID NO:41)
  • TABLE 2
    Additional results of shock experiments
    NMPF SEQUENCE ID:
    ANTI-SHOCK EFFECT
    LQGV (SEQ ID NO:1) +++
    AQGV (SEQ ID NO:2) +++
    LQGA (SEQ ID NO:19) +++
    VLPALP (SEQ ID NO:3) +++
    ALPALP (SEQ ID NO:21) ++
    VAPALP (SEQ ID NO:22) ++
    ALPALPQ (SEQ ID NO:23) ++
    VLPAAPQ (SEQ ID NO:24) ++
    VLPALAQ (SEQ ID NO:25) +++
    SHOCK ACCELERATING EFFECT
    LAGV (SEQ ID NO:26) +++
    LQAV (SEQ ID NO:52) +++
    VLAALP (SEQ ID NO:27) +++
    VLPAAP (SEQ ID NO:117) +++
    VLPALA (SEQ ID NO:28) +++
    VLPALPQ (SEQ ID NO:29) +++
    VLAALPQ (SEQ ID NO:30) +++
    VLPALPA (SEQ ID NO:31) +++
  • TABLE 3
    Further additional results of shock experiments
    % SURVIVAL IN TIME (HOURS)
    NMPF Tx Tx
    PEPTIDES 0 14 24 48
    PBS 100 100 100 0
    NMPF-3 100 100 100 0
    NMPF-5 100 100 100 100
    NMPF-7 100 100 100 67
    NMPF-8 100 100 100 100
    NMPF-9 100 100 100 100
    NMPF-11 100 100 100 100
    NMPF-12 100 100 100 100
    NMPF-43 100 100 100 100
    NMPF-45 100 100 100 100
    NMPF-46 100 100 100 100
    NMPF-50 100 100 100 100
    NMPF-53 100 100 100 100
    NMPF-58 100 100 100 100
    NMPF-60 100 100 100 100
  • TABLE 4
    Further additional results
    SICKNESS SCORES
    NMPF Tx Tx
    PEPTIDES 0 14 24 48
    PBS 0, 0, 0, 0, 0, 0 5, 5, 5, 5, 4, 4 5, 5, 5, 5, 5, 5 ††††††
    NMPF-3 0, 0, 0, 0, 0, 0 3, 3, 3, 3, 3, 4 4, 4, 4, 4, 4, 4 ††††††
    NMPF-5 0, 0, 0, 0, 0, 0 5, 5, 5, 5, 5, 5 5, 5, 5, 5, 5, 5 2, 2, 2, 2, 2, 2
    NMPF-7 0, 0, 0, 0, 0, 0 1, 1, 4, 4, 4, 4 5, 5, 5, 5, 5, 5 2, 2, 2, 2, ††
    NMPF-8 0, 0, 0, 0, 0, 0 3, 3, 5, 5, 5, 5 5, 5, 5, 5, 5, 5 2, 2, 4, 4, 4, 5
    NMPF-9 0, 0, 0, 0, 0, 0 3, 3, 4, 4, 5, 5 2, 2, 2, 2, 2, 2 1, 1, 2, 2, 2, 2
    NMPF-11 0, 0, 0, 0, 0, 0 1, 1, 3, 3, 4, 4, 2, 2, 2, 2, 4, 4 1, 1, 1, 1, 1, 1
    NMPF-12 0, 0, 0, 0, 0, 0 1, 1, 1, 1, 3, 3 1, 1, 1, 1, 1, 1 1, 1, 1, 1, 1, 1
    NMPF-43 0, 0, 0, 0, 0, 0 1, 1, 4, 4, 4, 4 1, 1, 1, 1, 3, 3 2, 2, 2, 2, 2, 2
    NMPF-45 0, 0, 0, 0, 0, 0 5, 5, 5, 5, 4, 4 3, 3, 4, 4, 5, 5 2, 2, 4, 4, 5, 5
    NMPF-46 0, 0, 0, 0, 0, 0 1, 1, 2, 2, 3, 3 1, 1, 2, 2, 2, 2 1, 1, 1, 1, 1, 1
    NMPF-50 0, 0, 0, 0, 0, 0 1, 1, 1, 1, 3, 3 2, 2, 2, 2, 3, 3 1, 1, 1, 1, 1, 1
    NMPF-53 0, 0, 0, 0, 0, 0 5, 5, 5, 5, 5, 5 5, 5, 5, 5, 5, 5 1, 1, 2, 2, 2, 2
    NMPF-58 0, 0, 0, 0, 0, 0 5, 5, 5, 5, 3, 3 5, 5, 5, 5, 3, 3 1, 1, 1, 1, 1, 1
    NMPF-60 0, 0, 0, 0, 0, 0 1, 1, 4, 4, 2, 2 2, 2, 2, 2, 4, 4 1, 1, 1, 1, 1, 1
  • TABLE 5
    Summary of results of the various peptides
    in the various experiments.
    ANGIO-
    ID SEQUENCE SEPSIS GENSIS CAO DC NOD
    NMPF-1 VLPALPQVVC −+ + +
    (SEQ ID NO:20)
    NMPF-2 LQGVLPALPQ −+ +
    (SEQ ID NO:49)
    NMPF-3 LQG −+ + + +
    NMPF-4 LQGV + + + +
    (SEQ ID NO:1)
    NMPF-5 GVLPALPQ −+ +
    (SEQ ID NO:33)
    NMPF-6 VLPALP + + + +
    (SEQ ID NO:3)
    NMPF-7 VLPALPQ + + +
    (SEQ ID NO:29)
    NMPF-8 GVLPALP −+ +
    (SEQ ID NO:32)
    NMPF-9 VVC + + +
    NMPF-10 QVVC
    (SEQ ID NO:43)
    NMPF-11 MTRV + + + +
    (SEQ ID NO:42)
    NMPF-12 MTR −+ + +
    NMPF-13 LQGVLPALPQVVC + +
    (SEQ ID NO:34)
    NMPF-14 cyclic-LQGVLPAL +
    PQVVC
    (SEQ ID NO:34)
    NMPF-43 AQG + + +
    NMPF-44 LAG +
    NMPF-45 LQA + +
    NMPF-46 AQGV + + +
    (SEQ ID NO:2)
    NMPF-47 LAGV −+ + +
    (SEQ ID NO:26)
    NMPF-48 LQAV
    (SEQ ID NO:52)
    NMPF-49 LQGA +
    (SEQ ID NO:19)
    NMPF-50 ALPALP + +
    (SEQ ID NO:21)
    NMPF-51 VAPALP + +
    (SEQ ID NO:22)
    NMPF-52 VLAALP
    (SEQ ID NO:27)
    NMPF-53 VLPAAP + +
    (SEQ ID NO:117)
    NMPF-54 VLPALA
    (SEQ ID NO:28)
    NMPF-55 ALPALPQ +
    (SEQ ID NO:23)
    NMPF-56 VAPALPQ +
    (SEQ ID NO:173)
    NMPF-57 VLAALPQ
    (SEQ ID NO:30)
    NMPF-58 VLPAAPQ + +
    (SEQ ID NO:24)
    NMPF-59 VLPALAQ + +
    (SEQ ID NO:25)
    NMPF-60 VLPALPA + +
    (SEQ ID NO:31)
    NMPF-61 VVCNYRDVRFESIRL −+ +
    PGCPRGVNPVVSYAV
    ALSCQCAL
    (SEQ ID NO:35)
    NMPF-62 VVCNYRDVRFESIRL
    PGCPRGVNPVVSYAV
    ALSCQ
    (SEQ ID NO:169)
    NMPF-63 SIRLPGCPRGVNP −+
    VVS
    (SEQ ID NO:39)
    NMPF-64 LPGCPRGVNPVVS +
    (SEQ ID NO:40)
    NMPF-65 CPRGVNPVVS
    (SEQ ID NO:50)
    NMPF-66 LPGC + + +
    (SEQ ID NO:41)
    NMPF-67 CPRGVNP
    (SEQ ID NO:170)
    NMPF-68 PGCP −+
    (SEQ ID NO:10)
    NMPF-69 RPRCRPINATLAVEK
    EGCPVCITVNTITLC
    AGYCPT
    (SEQ ID NO:45)
    NMPF-70 MTRVLQGVLPALPQ −+
    (SEQ ID NO:171)
    NMPF-71 MTRVLPGVLPALPQ −+
    VVC
    (SEQ ID NO:174)
    NMPF-74 CALCRRSTTDCGGPK
    DHPLTC
    (SEQ ID NO:46)
    NMPF-75 SKAPPPSLPSPSRLP
    GPC
    (SEQ ID NO:172)
    NMPF-76 TCDDPRFQDSSSSKA
    PPPSLPSPSRLPGPS
    DTPILPQ
    (SEQ ID NO:48)
    + = effects;
    −+ = variable effect;
    no entry is no effect or not yet tested when table was assembled
  • TABLE 6
    MODULATION OF NO AND/OR TNF-α
    ID SEQUENCE TNF-A NO TNF-A and NO
    NMPF-1 VLPALPQVVC ++ ++++ ++++
    (SEQ ID NO:20)
    NMPF-2 LQGVLPALPQ −+ ++++ ++++
    (SEQ ID NO:49)
    NMPF-3 LQG + ++++ ++++
    NMPF-4 LQGV ++++ ++++ +++++++
    (SEQ ID NO:1)
    NMPF-5 GVLPALPQ ++++ ++++ +++++++
    (SEQ ID NO:33)
    NMPF-6 VLPALP ++++ ++++ +++++++
    (SEQ ID NO:3)
    NMPF-7 VLPALPQ ++++ ++++ +++++++
    (SEQ ID NO:29)
    NMPF-8 GVLPALP ++++ ++++ +++++++
    (SEQ ID NO:32)
    NMPF-9 VVC ++++ ++++ +++++++
    NMPF-1O QVVC ++++ +++ ++++
    (SEQ ID NO:43)
    NMPF-11 MTRV ++++ ++++ ++++
    (SEQ ID NO:42)
    NMPF-12 MTR ++++ ++++ ++++
    NMPF-13 LQGVLPALPQVVC ++ ++++ ++++
    (SEQ ID NO:34)
    NMPF-14 cyclic-LQGVLPAL ++ ++++ ++++
    PQVVC
    (SEQ ID NO:34)
    NMPF-43 AQG ++++ ++++ +++++++
    NMPF-44 LAG −+ ++++ ++++
    NMPF-45 LQA ++++ ++++ +++++++
    NMPF-46 AQGV ++++ ++++ +++++++
    (SEQ ID NO:2)
    NMPF-47 LAGV ++ ++++ ++++
    (SEQ ID NO:26)
    NMPF-48 LQAV ++ ++++ ++++
    (SEQ ID NO:52)
    NMPF-49 LQGA ++ ++++ ++++
    (SEQ ID NO:19)
    NMPF-50 ALPALP ++++ ++++ +++++++
    (SEQ ID NO:21)
    NMPF-51 VALPALP + +++ ++++
    (SEQ ID NO:22)
    NMPF-52 VLAALP ++ ++++ ++++
    (SEQ ID NO:27)
    NMPF-53 VLPAAP ++++ ++++ +++++++
    (SEQ ID NO:117)
    NMPF-54 VLPALA + ++++ ++++
    (SEQ ID NO:28)
    NMPF-55 ALPALPQ + ++++ ++++
    (SEQ ID NO:23)
    NMPF-56 VALPALPQ −+ ++++ ++++
    (SEQ ID NO:173)
    NMPF-57 VLAALPQ + ++++ ++++
    (SEQ ID NO:30)
    NMPF-58 VLPAAPQ ++++ ++++ +++++++
    (SEQ ID NO:24)
    NMPF-59 VLPALAQ ++ ++++ ++++
    (SEQ ID NO:25)
    NMPF-60 VLPALPA ++++ ++++ +++++++
    (SEQ ID NO:31)
    NMPF-61 VVCNYRDVRFESIRL −+ ++++ ++++
    PGCPRGVNPVVSYAV
    ALSCQCAL
    (SEQ ID NO:35)
    NMPF-62 VVCNYRDVRFESIRL −+ +++ ++++
    PGCPRGVNPVVSYAV
    ALSCQ
    (SEQ ID NO:169)
    NMPF-63 SIRLPGCPRGVNP −+ ++ ++
    VVS
    (SEQ ID NO:39)
    NMPF-64 LPGCPRGVNPVVS ++ ++++ ++++
    (SEQ ID NO:40)
    NMPF-65 CPRGVNPVVS ++ +++ +++
    (SEQ ID NO:50)
    NMPF-66 LPGC +++ ++ +++
    (SEQ ID NO:41)
    NMPF-67 CPRGVNP −+ + +
    (SEQ ID NO:170)
    NMPF-68 PGCP + + +++
    (SEQ ID NO:10)
    NMPF-69 RPRCRPINATLAVEK −+ ++ ++
    EGCPVCITVNTTICA
    GYCPT
    (SEQ ID NO:45)
    NMPF-70 MTRVLQGVLPALPQ −+ + +
    (SEQ ID NO:171)
    NMPF-71 MTRVLPGVLPALPQV −+ −+ −+
    VC
    (SEQ ID NO:174)
    NMPF-74 CALCRRSTTDCGGPK −+ ++ +
    DHPLTC
    (SEQ ID NO:46)
    NMPF-75 SKAPPPSLPSPSRIP + ++ ++
    GPS
    (SEQ ID NO:172)
    NMPF-76 TCDDPRFQDSSSSKA + + +
    PPPSLPSPSRLPGPS
    DTPILPQ
    (SEQ ID NO:48)
    NMPF-78 CRRSTTDCGGPKDHP + + +
    LTC
    (SEQ ID NO:47)
    from −+ to +++++++ indicates from barely active to very active in modulating
  • Example V Monkey Experiment
  • Efficacy of NMPF, here a mixture 1:1:1 of LQGV (SEQ ID NO:1), AQGV (SEQ ID NO:2) and VLPALP (SEQ ID NO:3), administered in a gram-negative-induced rhesus monkey sepsis model for prevention of septic shock.
  • Overwhelming inflammatory and immune responses are essential features of septic shock and play a central part in the pathogenesis of tissue damage, multiple organ failure, and death induced by sepsis. Cytokines, especially tumor necrosis factor (TNF)-α interleukin (IL)-1β, and macrophage migration inhibitory factor (MIF), have been shown to be critical mediators of septic shock. Yet, traditional anti-TNF and anti-IL-1 therapies have not demonstrated much benefit for patients with severe sepsis. We have designed peptides that block completely LPS-induced septic shock in mice, even when treatment with these peptides is started up to 24 hours after LPS injection. These peptides are also able to inhibit the production of MIF. This finding provides the possibility of therapeutic use of these peptides for the treatment of patients suffering from septic shock. Since primates are evolutionary closer to humans, we tested these peptides for their safety and effectiveness in a primate system.
  • EXPERIMENTAL DESIGN
    EXPERIMENTAL
    TREATMENT
    (independent variable,
    e.g., placebo treated
    GROUP control group) BIOTECHNIQUES NUMBER
    animal I i.v. infusion of a lethal Live E. coli infusion N = 1
    dose of live Escherichia Blood sampling
    coli (10E10 CFU/kg) + No recovery (section)
    antibiotics +
    placebo treated
    animal II i.v. infusion of a lethal Live E. coli infusion N = 1
    dose of live Escherichia Blood sampling
    coli (10E10 CFU/kg) + No recovery (section)
    antibiotics +
    oligopeptide (5 mg/kg
    of each of 3 peptides)
  • Only naive monkeys were used in this preclinical study to exclude any interaction with previous treatments. The animals were sedated with ketamine hydrochloride. Animals were intubated orally and allowed to breathe freely. The animals were kept anesthetized with O2/N2O/isoflurane. The animals received atropine as pre-medication for O2/N2O/isoflurane anesthesia. A level of surgical anesthesia was maintained during the two hours infusion of E. coli and for six hours following E. coli challenge, after which the endotracheal tubes were removed and the animals were euthanized. Before bacteria were induced, a one hour pre-infusion monitoring of heart-rate and blood pressure was performed.
  • Two rhesus monkeys were infused with a 1010 CFU per kg of the gram-negative bacterium E. coli to induce a fatal septic shock. One monkey received placebo-treatment and was sacrificed within seven hours after infusion of the bacteria without recovery from the anesthesia. The second monkey received treatment with test compound and was sacrificed at the same time point.
  • In a limited dose-titration experiment performed with the same bacterium strain in 1991, the used dose proved to induce fatal shock within eight hours. In recent experiments, a three-fold lower dose was used inducing clear clinical and pathomorphological signs of septic shock without fatal outcome.
  • The monkeys were kept anesthetized throughout the observation period and sacrificed seven hours after the start of the bacterium infusion for pathological examination. The animals underwent a gross necropsy in which the abdominal and thorax cavities were opened and internal organs examined in situ.
  • Full Description of the Experiment with Three Rhesus Monkeys
  • The study was conducted in rhesus monkeys (Maccaca mulatta). Only experimentally naive monkeys were used in the study to exclude any interaction with previous treatments. Prior to the experiment, the state of health of the animals was assessed physically by a veterinarian. All animals had been declared to be in good health and were free of pathogenic ecto- and endoparasites and common bacteriological infections: Yersinia pestis, Yersinia enterocolitica, Yersinia pseudotuberculosis, Shigella, Aeromonas hydrophilia, pathogenic Campylobacter species and Salmonella.
  • Reagents. The Escherichia coli strain was purchased from ATCC (E. coli; 086a: K61 serotype, ATCC 33985). In a control experiment, the strain proved equally susceptible to bactericidal factors in human and rhesus monkey serum. Prior to the experiment, a fresh culture was set-up; the E. coli strain was cultured for one day, harvested and washed thoroughly to remove free endotoxin. Prior to infusion into the animal, the number and viability of the bacteria were assessed. Serial dilutions of the E. coli stock were plated on BHI agar and cultured overnight at 37° C. The colonies on each plate were counted and the number of colony-forming units per ml was calculated. The body weight measurement of the day of the experiment was used to calculate the E. coli dose and E. coli stock was suspended in isotonic saline (N.P.B.I., Emmer-Compascuum, NL) at the concentration needed for infusion (total dose volume for infusion approximately 10 ml/kg. The E. coli suspension was kept on ice until infusion.
  • Antibiotic was used to synchronize the shock induction in the monkeys. Baytril (Baytril 2.5%, Bayer, Del.) was used instead of gentamycin, as the strain proved only marginally susceptible to the latter antibiotic. Individual animals were identified by a number or letter combination tattooed on the chest.
  • Experimental design.
    GROUP
    (number/ EXPERIMENTAL
    letter or TREATMENT
    other (independent variable,
    identi- e.g., placebo treated
    fication control group) NUMBER SEX
    Animal i.v. infusion of a lethal Live E. coli N = 1 F
    I dose of live Escherichia infusion
    coli (10E10 CFU/kg) + Blood sampling
    antibiotic + No recovery
    placebo treated
    Animal i.v. infusion of a lethal Live E. coli N = 1 F
    II dose of live Escherichia infusion
    coli (10E10 CFU/kg) + Blood sampling
    antibiotic + No recovery
    NMPF-4, -6, -46; each (section)
    5 mg/kg
    Animal i.v. infusion of a lethal Live E. coli N = 1 F
    III dose of live Escherichia infusion
    coli (10E10 CFU/kg) + Blood sampling
    antibiotic + Recovery and
    NMPF-4, -6, -46; each survival
    5 mg/kg
  • Anesthesia. All animals were fasted overnight prior to the experiment. On the morning of the experiment, the animals were sedated with ketamine hydrochloride (Tesink, NL) and transported to the surgery. The animal was placed on its side on a temperature-controlled heating pad to support body temperature. Rectal temperature was monitored using a Vet-OX 5700. The animals were intubated orally and were allowed to breathe freely. The animals were kept anesthetized using O2/N2O/isoflurane inhalation anesthesia during the E. coli infusion and the seven-hour observation period following E. coli challenge, after which the endotracheal tubes were removed and the animals were euthanized or allowed to recover from anesthesia. The femoral or the cephalic vein was cannulated and used for infusing isotonic saline, live E. coli and antibiotic administration. Insensible fluid loss was compensated for by infusing isotonic saline containing 2.5% glucose (Fresenius, 's Hertogenbosch, NL) at a rate of 3.3 ml/kg/hour.
  • Preparative actions. During anesthesia the animals were instrumented for measurement of blood pressure (with an automatic cuff), heart rate and body temperature. Isotonic saline was infused at 3.3 ml/kg/hour to compensate for fluid loss. Femoral vessels were cannulated for infusion of E. coli and antibiotics. Temperature-controlled heating pads were used to support body temperature. The monkeys were continuously monitored during the E. coli challenge and for the six-hour period following E. coli administration. After seven hours, two animals (the control animal and one treated with NMPF) were sacrificed to compare the direct effect of the compound at the level of histology. The third animal, treated with NMPF, was allowed to recover from anesthesia and was intensively observed during the first 12 hours after recovery followed by frequent daily observation. The decision to allow the third animal to recover was made after consulting with the veterinarian.
  • Induction of septic shock. Before the infusion of E. coli, a one-hour pre-infusion monitoring of heart-rate and blood pressure was performed. All three animals received an i.v. injection of E. coli 086 (k61 serotype; ATCC 33985) at a lethal dose of 10×109 CFU/kg body weight. In a dose titration study with this batch performed in 1991, this bacterial dose induced lethal shock within eight hours after the start of the infusion. The infusion period was two hours.
  • Antibiotics. Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg).
  • Treatment with NMPF. 30 minutes post-onset of E. coli infusion, the animals were administered a single intravenous bolus injection of a mixer of NMPF oligopeptides. The oligopeptide mixer contained the following NMPF peptides: LQGV (SEQ ID NO:1) (5 mg/kg), AQGV (SEQ ID NO:2) (5 mg/kg) and VLPALP (SEQ ID NO:3) (5 mg/kg). These NMPF peptides were dissolved in 0.9% sodium chloride for injection (N.P.B.I., Emmer Compascuum, NL).
  • Results Preliminary Monkey Results
  • An anti-shock effect of the test compound on sepsis in the monkey treated with the oligopeptide mixture, namely the inhibition of the effect of the sepsis in this early seven-hour trajectory of this primate model, was observed. Immunomodulatory effects with these peptides have been observed in vitrolex vivo such as in T-cell assays, the inhibition of pathological Th1 immune responses, suppression of inflammatory cytokines (MIF), increase in production of anti-inflammatory cytokines (IL-10, TGF-beta) and immunomodulatory effects on antigen-presenting cells (APC) like dendritic cells and macrophages.
  • The following organs were weighed and a bacterial count was performed: kidneys, liver, lungs, lymph nodes, and gross lesions.
  • Tissues of all organs were preserved in neutral aqueous phosphate buffered 4% solution of formaldehyde. Lymphoid organs were cryopreserved. All tissues will be processed for histopathological examination.
  • Further Results Obtained in the Three-Monkey Experiment
  • Monkey 429(control). Female monkey (5.66 kg) received an i.v. injection of E. coli 086 (1010 CFU/kg). In a dose titration study with this batch performed in 1991, this bacterial dose induced lethal shock within eight hours after the start of the infusion. The infusion period was two hours. Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg). After the E. coli injection, the monkey was observed by the authorized veterinarian without knowing which of the monkeys received NMPF treatment. The clinical observations were as follows: vomiting, undetectable pulse, heart arrhythmia, abnormalities in ECG: signs of ventricle dilatation/heart decompensation (prolonged QRS complex, extra systoles), decreased blood clotting and forced respiration. In addition, there was big fluctuation in heart rate (30-150 beats per minute), collapse of both systolic and diastolic blood pressure (35/20 mmHg) and decrease in blood oxygen concentration (80-70%). Seven hours after the start of the E. coli infusion, monkey began to vomit blood and feces, and have convulsions. After final examination, the veterinarian did not give permission to let this monkey awake. At this time point, the control monkey was euthanized. Hereafter, post-mortem examination was conducted and internal organs were examined in situ. A number of internal bleedings were found by the pathologist.
  • Monkey 459 (NMPF). Female monkey (5.44 kg) received an i.v. injection of E. coli 086 (1010 CFU/kg). In a dose titration study with this batch performed in 1991, this bacterial dose induced lethal shock within eight hours after the start of the infusion. The infusion period was two hours. Thirty minutes after the initiation of E. coli infusion, NMPF was i.v. injected in a single bolus injection. Baytril was administered intravenously immediately after completion of the two-hour E. coli infusion (i.v.; dose 9 mg/kg). After the E. coli injection, this monkey was also observed by the authorized veterinarian without knowing which of the monkeys received NMPF treatment. The clinical observations were as follows: normal pulse, heart sounds normal, normal ECG, higher heart-rate but otherwise stable (180 beats per minute), no hypotension (75/30 mmHg), normal blood oxygen concentration (95-85%), lungs sound normal, normal turgor. Seven hours after the start of the E. coli infusion, the clinical condition of the monkey was stable. After final examination, the veterinarian did give permission to let this monkey awake due to her stable condition. In order to compare the hematological and immunological parameters between the control and NMPF-treated monkey, at this time point the NMPF-treated monkey 459 was euthanized. Hereafter, post-mortem examination was conducted and internal organs were examined in situ. No macroscopic internal bleedings were found by the pathologist.
  • Monkey 427(NMPF). Female monkey (4.84 kg) received an i.v. injection of E. coli 086 (1010 CFU/kg). In a dose titration study with this batch performed in 1991, this bacterial dose induced lethal shock within eight hours after the start of the infusion. The infusion period was two hours. Thirty minutes after the initiation of E. coli infusion, NMPF was i.v. injected. Baytril was administered intravenously immediately after completion of the two hours. E. coli infusion (i.v.; dose 9 mg/kg). After the E. coli injection, this monkey was also observed by the authorized veterinarian doctor without knowing which of the monkeys received NMPF treatment. The clinical observations were as follows: normal pulse, heart sounds normal, normal ECG, moderately higher heart-rate but otherwise stable (160 beats per minute), no hypotension (70/30 mmHg), normal blood oxygen concentration (95-90%), lungs sound normal, normal turgor. Seven hours after the start of the E. coli infusion, the clinical condition of the monkey was stable. After final examination, the veterinarian did give permission to let this monkey wake up due to her stable condition. The monkey woke up quickly, she was alert, and there was a slow disappearance of oedema.

Claims (4)

1. A composition comprising one or more purified or isolated peptides of the sequence LAGV (SEQ ID NO:26) or salt thereof.
2. A composition comprising a purified or isolated peptide of the sequence LAGV (SEQ ID NO:26) or salt thereof.
3. A composition comprising:
a purified peptide consisting of the sequence of SEQ ID NO:26, or a salt thereof.
4. The composition of claim 3, further comprising:
a pharmaceutically acceptable carrier, diluent and/or excipient.
US12/069,401 2001-03-29 2008-02-08 Immunoregulatory compositions Abandoned US20080306009A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/069,401 US20080306009A1 (en) 2001-03-29 2008-02-08 Immunoregulatory compositions

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
PCT/NL2001/000259 WO2001072831A2 (en) 2000-03-29 2001-03-29 Immunoregulator
US10/028,075 US20030113733A1 (en) 2001-10-04 2001-12-21 Gene regulator
US10/262,522 US7365155B2 (en) 2000-03-29 2002-09-30 Immunoregulator
PCT/NL2002/000639 WO2003029292A2 (en) 2001-10-04 2002-10-04 Gene regulatory peptides
US10/753,510 US7358330B2 (en) 2001-03-29 2004-01-07 Immunoregulatory compositions
US12/069,401 US20080306009A1 (en) 2001-03-29 2008-02-08 Immunoregulatory compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/753,510 Division US7358330B2 (en) 2000-03-29 2004-01-07 Immunoregulatory compositions

Publications (1)

Publication Number Publication Date
US20080306009A1 true US20080306009A1 (en) 2008-12-11

Family

ID=46300652

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/753,510 Ceased US7358330B2 (en) 2000-03-29 2004-01-07 Immunoregulatory compositions
US11/859,265 Abandoned US20080076719A1 (en) 2001-03-29 2007-09-21 Immunoregulatory compositions
US12/069,401 Abandoned US20080306009A1 (en) 2001-03-29 2008-02-08 Immunoregulatory compositions
US13/065,317 Expired - Lifetime USRE43309E1 (en) 2000-03-29 2011-03-17 Immunoregulatory compositions

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/753,510 Ceased US7358330B2 (en) 2000-03-29 2004-01-07 Immunoregulatory compositions
US11/859,265 Abandoned US20080076719A1 (en) 2001-03-29 2007-09-21 Immunoregulatory compositions

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/065,317 Expired - Lifetime USRE43309E1 (en) 2000-03-29 2011-03-17 Immunoregulatory compositions

Country Status (1)

Country Link
US (4) US7358330B2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070054860A1 (en) * 2001-12-21 2007-03-08 Biotempt B.V. Treatment of ischemic events
US20100004172A1 (en) * 2003-04-08 2010-01-07 Khan Nisar A Compositions for mucosal and oral administration comprising hcg fragments
US7989160B2 (en) 2006-02-13 2011-08-02 Alethia Biotherapeutics Inc. Polynucleotides and polypeptide sequences involved in the process of bone remodeling
USRE43309E1 (en) 2000-03-29 2012-04-10 Biotempt B.V. Immunoregulatory compositions
US8168181B2 (en) 2006-02-13 2012-05-01 Alethia Biotherapeutics, Inc. Methods of impairing osteoclast differentiation using antibodies that bind siglec-15
US8288341B2 (en) 2006-03-07 2012-10-16 Biotempt B.V. Control of radiation injury
US9493562B2 (en) 2012-07-19 2016-11-15 Alethia Biotherapeutics Inc. Anti-Siglec-15 antibodies

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6921751B1 (en) * 1998-05-20 2005-07-26 Erasmus Universiteit Rotterdam Immunoregulator
US6844315B2 (en) * 1998-05-20 2005-01-18 Erasmus Universiteit Rotterdam Immunoregulator
US20040202645A1 (en) * 2003-04-08 2004-10-14 Khan Nisar Ahmed Administration of gene-regulatory peptides
EP1138692A1 (en) 2000-03-29 2001-10-04 Erasmus Universiteit Rotterdam Fragments of human chorionic gonadotropin (hcg) as immunoregulator
US8680059B2 (en) * 1998-05-20 2014-03-25 Biotempt B.V. Oligopeptide acetate and formulations thereof
US20050227925A1 (en) * 2004-04-08 2005-10-13 Robbert Benner Compositions capable of reducing elevated blood urea concentration
USRE43279E1 (en) 2000-03-29 2012-03-27 Biotemp B.V. Compositions capable of reducing elevated blood urea concentration
EP1300418A1 (en) * 2001-10-04 2003-04-09 Erasmus Universiteit Rotterdam Gene regulation by oligopeptides
US20030220259A1 (en) * 2001-12-21 2003-11-27 Robbert Benner Treatment of neurological disorders
US20080318871A1 (en) * 2001-12-21 2008-12-25 Khan Nisar A Treatment of neurological disorders
US20030220257A1 (en) * 2001-12-21 2003-11-27 Robbert Benner Treatment of trauma
US7501391B2 (en) * 2001-12-21 2009-03-10 Biotempt B.V. Treatment of transplant survival
US7560433B2 (en) 2001-12-21 2009-07-14 Biotempt B.V. Treatment of multiple sclerosis (MS)
US20030224995A1 (en) * 2001-12-21 2003-12-04 Khan Nisar Ahmed Treatment of burns
US20030220260A1 (en) * 2001-12-21 2003-11-27 Khan Nisar Ahmed Peptide compositions
US7786084B2 (en) * 2001-12-21 2010-08-31 Biotempt B.V. Treatment of burns
US20040013661A1 (en) * 2001-12-21 2004-01-22 Gert Wensvoort Stratification
US20080242837A1 (en) * 2001-12-21 2008-10-02 Khan Nisar A Peptide compositions
US20030220261A1 (en) * 2001-12-21 2003-11-27 Khan Nisar Ahmed Treatment of iatrogenic disease
US20090227505A1 (en) * 2004-01-07 2009-09-10 Biotempt B.V. Methods and uses for protein breakdown products
JP4910700B2 (en) * 2004-08-30 2012-04-04 東レ株式会社 Fractionation device
SG163567A1 (en) * 2005-07-05 2010-08-30 Biotempt Bv Treatment of tumors
WO2007097624A1 (en) * 2006-02-21 2007-08-30 Biotempt B.V. Pharmaceutical compositions for the treatment of influenza infections
PL2120991T3 (en) * 2007-02-12 2014-07-31 Biotempt Bv Treatment of trauma hemorrhage with short oligopeptides
WO2023059188A1 (en) 2021-10-05 2023-04-13 Biotempt B.V. Angiogenic control, preferably combined with glycaemic control.

Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3855397A (en) * 1973-04-12 1974-12-17 Allied Chem Method of producing sodium carbonate and bicarbonate spherules from brine
US3898328A (en) * 1973-12-03 1975-08-05 Syntex Inc Dry stable composition for the treatment of scours and dehydration
US3960830A (en) * 1973-12-06 1976-06-01 Hoechst Aktiengesellschaft Polyalkylene glycols used for the preparation of peptides
US4003989A (en) * 1974-05-06 1977-01-18 Bar On Ernest Pharmaceutical preparation
US4083951A (en) * 1976-05-05 1978-04-11 Beecham Group Limited Pharmaceutical composition
US4108846A (en) * 1977-02-01 1978-08-22 Hoffmann-La Roche Inc. Solid phase synthesis with base N alpha-protecting group cleavage
US4140760A (en) * 1976-11-09 1979-02-20 Reckitt & Colman Products Limited Pharmaceutical compositions for use in the suppression of gastric reflux
US4265874A (en) * 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4289750A (en) * 1978-10-16 1981-09-15 Kopp Klaus F Therapy of conditions which may be associated with altered renal function and dosage forms therefor
US4351762A (en) * 1981-03-10 1982-09-28 Bioresearch, Inc. Rapid, quantitative peptide synthesis using mixed anhydrides
US5070187A (en) * 1989-11-03 1991-12-03 Trustees Of Boston University Pharmacologically effective antagonists of arginine-vasopressin
US5137669A (en) * 1988-03-02 1992-08-11 Endocon, Inc. Manufacture of partially fused peptide pellet
US5175254A (en) * 1988-09-24 1992-12-29 Societe D'expansion Scientifque Expansia Solid phase peptide synthesis using a polyacrylic support in aqueous solution
US5200507A (en) * 1991-04-12 1993-04-06 Mallinckrodt Specialty Chemicals Company Method of separating a peptide from a resin
US5554378A (en) * 1990-09-03 1996-09-10 Takeda Chemical Industries, Ltd. Pharmaceutical composition and its mucosal use
US5595760A (en) * 1994-09-02 1997-01-21 Delab Sustained release of peptides from pharmaceutical compositions
US5610272A (en) * 1988-05-10 1997-03-11 Alpha-1 Biomedicals, Inc. Solid phase process for synthesizing thymosin α1
US5616608A (en) * 1993-07-29 1997-04-01 The United States Of America As Represented By The Department Of Health And Human Services Method of treating atherosclerosis or restenosis using microtubule stabilizing agent
US5639480A (en) * 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5665394A (en) * 1994-02-21 1997-09-09 Takeda Chemical Industries, Ltd. Matrix for sustained-release preparation
US5668111A (en) * 1992-12-07 1997-09-16 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5837218A (en) * 1995-09-15 1998-11-17 Resolution Pharmaceuticals Inc. Non-receptor cell mediated imaging agents
US5858375A (en) * 1994-09-15 1999-01-12 Medeva Europe Limited Pharmaceutical compositions containing D2 O
US20030044463A1 (en) * 2001-09-06 2003-03-06 Romano Deghenghi Sustained release of microcrystalline peptide suspensions
US20030229022A1 (en) * 2002-06-07 2003-12-11 Fortuna Haviv Pharmaceutical formulation
US20080267967A1 (en) * 2001-05-24 2008-10-30 Gorczynski Reginald M Modulation of cd200 receptors
US20090093398A1 (en) * 2003-11-07 2009-04-09 Jacques Bollekens Use of Fibroblast Growth Factor Fragments
US7786084B2 (en) * 2001-12-21 2010-08-31 Biotempt B.V. Treatment of burns
US7795226B2 (en) * 2006-03-07 2010-09-14 Biotempt B.V. Control of radiation injury
US20110105415A1 (en) * 2001-10-04 2011-05-05 Biotempt B.V. Gene regulator
US20110113053A1 (en) * 2008-04-09 2011-05-12 Nisar Ahmed Khan Methods for identifying biologically active peptides and predicting their function

Family Cites Families (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5822474B2 (en) * 1980-06-10 1983-05-09 天野製薬株式会社 Method for producing cholecystokinin pancreozymin C-terminal peptide amide sulfate ester
US4427660A (en) 1982-03-03 1984-01-24 Research Corporation Formyl-methionyl chemotatic peptide antibiotic conjugates useful in treating infections
US4571336A (en) 1983-08-25 1986-02-18 Endorphin, Inc. Immune stimulation
US4977244A (en) 1985-06-27 1990-12-11 The United States Of America As Represented By The Department Of Health And Human Services Uromodulin and a process of purifying it
US4855285A (en) 1985-12-04 1989-08-08 The Ohio State University Research Foundation Antigenic modification of polypeptides
DE3715662A1 (en) 1986-05-12 1987-11-19 Wellcome Found NEW PHARMACEUTICAL APPLICATION
US4753965A (en) 1987-04-09 1988-06-28 Merrell Dow Pharmaceuticals, Inc. Method of treating multiple sclerosis with chalcone derivatives
US5002961A (en) 1987-10-19 1991-03-26 Merrell Dow Pharmaceuticals Method for reducing injury with imidazol-2-thionecarboxamides
US5055447A (en) * 1988-07-28 1991-10-08 Genentech, Inc. Method and compositions for the treatment and prevention of septic shock
US5308834A (en) 1989-02-14 1994-05-03 Incyte Pharmaceuticals, Inc. Treatment of endotoxin-associated shock and prevention thereof using a BPI protein
US6051596A (en) 1990-08-10 2000-04-18 Anormed, Inc. Immunosuppressive compositions
US5958413A (en) 1990-11-01 1999-09-28 Celltech Limited Use of antibodies to TNF or fragments derived thereof and xanthine derivatives for combination therapy and compositions therefor
EP0584238A1 (en) 1991-05-17 1994-03-02 Chiron Corporation INHIBITOR OF NF-$g(k)B TRANSCRIPTIONAL ACTIVATOR AND USES THEREOF
US5223397A (en) 1991-06-05 1993-06-29 Sangstat Medical Corporation Soluble hla cross-match
DE4138621A1 (en) 1991-11-25 1993-06-17 Boehringer Ingelheim Int METHOD FOR SCREENING SUBSTANCES WITH MODULATING EFFECT ON A RECEPTACLE-RELATED CELLULAR SIGNAL TRANSMISSION PATH
ATE260971T1 (en) 1992-04-01 2004-03-15 Univ Rockefeller METHOD FOR THE IN VITRO CULTIVATION OF DENDRITIC PRECURSOR CELLS AND THEIR USE FOR IMMUNOGENIC PRODUCTION
DE69219590T2 (en) 1992-05-30 1997-11-27 Sikiric Predrag BPC peptides, their production and use
US5436270A (en) 1993-04-07 1995-07-25 National Science Council Method for protecting against endotoxin-induced shock
FR2706772A1 (en) 1993-06-22 1994-12-30 Vacsyn Sa Prevention and treatment of septic syndrome with an immunosuppressant, in particular cyclosporin.
US5380668A (en) 1993-07-06 1995-01-10 University Of Utah Research Foundation Compounds having the antigenicity of hCG
US5837478A (en) * 1993-12-23 1998-11-17 Icos Corporation Method of identifying modulators of binding between and VCAM-1
NZ278607A (en) 1994-02-07 1999-05-28 Knoll Ag Use of tnf antagonists for treating disorders involving elevated serum levels of il-6 wherein the serum levels are 500pg/ml or above
US5677275A (en) 1994-08-05 1997-10-14 The United States Of America As Represented By The Department Of Health And Human Services Treatment of cancer with human chorionic gonadotropin
US5851997A (en) 1994-10-04 1998-12-22 Harris; Pamela Jo Use of human chorionic gonadotropin as an immune-potentiating antiviral agent
US5700781A (en) 1994-10-04 1997-12-23 Harris; Pamela Jo Method for treating Kaposi's sarcoma and HIV infections
SE520730C2 (en) 1995-01-20 2003-08-19 Eskil Elmer Treatment of brain ischemia and brain damage with a neuroprotective drug
EP0867448A4 (en) 1995-04-13 2000-04-26 Chugai Pharmaceutical Co Ltd PEPTIDE SUPPRESSING IxBalpha PHOSPHORYLATION
AU5161296A (en) 1995-04-21 1996-11-07 Allelix Biopharmaceuticals Inc. Anti-haemorrhagic peptides
US5942494A (en) 1995-10-06 1999-08-24 The Trustees Of Columbia University In The City Of New York Stimulation of gene expression and synthesis of heat shock protein 72/73 (HSP 70)
JPH09176187A (en) 1995-12-27 1997-07-08 Kagaku Gijutsucho Hoshasen Igaku Sogo Kenkyusho Histidine-containing peptide having active oxygen scavenging activity
US6361992B1 (en) 1996-05-08 2002-03-26 The United States Of America As Represented By The Department Of Health And Human Services Thyroid stimulating hormone superagonists
WO1997049721A1 (en) 1996-06-24 1997-12-31 University Of Maryland Biotechnology Institute Methods of treatment of wasting syndrome based on administration of derivatives of human chorionic gonadotropin
US5968513A (en) 1996-06-24 1999-10-19 University Of Maryland Biotechnology Institute Method of promoting hematopoiesis using derivatives of human chorionic gonadotropin
US5997871A (en) 1996-06-24 1999-12-07 University Of Maryland Biotechnology Insitute Treatment and prevention of cancer by administration of derivatives of human chorionic gonadotropin
US6319504B1 (en) 1996-06-24 2001-11-20 University Of Maryland Biotechnology Institute Treatment and prevention of HIV infection by administration of derivatives of human chorionic gonadotropin
CA2180786C (en) * 1996-07-02 2008-05-27 David Lightfoot Plants containing the gdha gene and methods of use thereof
US6150500A (en) 1996-07-12 2000-11-21 Salerno; John C. Activators of endothelial nitric oxide synthase
GB9617021D0 (en) 1996-08-13 1996-09-25 Salpep Biotechnology Inc Novel peptides for treatment of inflammation and shock
GB9624927D0 (en) * 1996-11-29 1997-01-15 Oxford Glycosciences Uk Ltd Gels and their use
US5966712A (en) * 1996-12-12 1999-10-12 Incyte Pharmaceuticals, Inc. Database and system for storing, comparing and displaying genomic information
US6346602B1 (en) 1997-02-07 2002-02-12 Thomas Jefferson University Peptide mimics of the cytokine receptor common γ chain and methods and compositions for making and using the same
AU741802B2 (en) 1997-02-15 2001-12-13 Millennium Pharmaceuticals, Inc. Treatment of infarcts through inhibition of NF-kappaB
US6416959B1 (en) * 1997-02-27 2002-07-09 Kenneth Giuliano System for cell-based screening
US5801193A (en) 1997-04-15 1998-09-01 Immune Modulation, Inc. Compositions and methods for immunosuppressing
US6583109B1 (en) 1997-06-24 2003-06-24 Robert C. Gallo Therapeutic polypeptides from β-hCG and derivatives
DE19735587B4 (en) 1997-08-16 2012-03-22 Eberhard-Karls-Universität Tübingen Universitätsklinikum A peptide having a radioprotective effect, a cosmetic or pharmaceutical composition containing the same, a nucleic acid encoding the same, a preparation process for this peptide and the use as a radioprotective agent
HUP0001237A3 (en) 1997-10-20 2002-01-28 Lilly Co Eli Methods for treating vascular disorders
US6831057B2 (en) 1997-10-28 2004-12-14 The University Of North Carolina At Chapel Hill Use of NF-κB inhibition in combination therapy for cancer
ES2276481T3 (en) 1997-12-12 2007-06-16 The University Of Western Ontario PEPTIDO APOEP1.B NOVEDOSO, COMPOSITIONS AND ITS USES.
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
US20040202645A1 (en) 2003-04-08 2004-10-14 Khan Nisar Ahmed Administration of gene-regulatory peptides
US8680059B2 (en) 1998-05-20 2014-03-25 Biotempt B.V. Oligopeptide acetate and formulations thereof
JP4804626B2 (en) 1998-05-20 2011-11-02 バイオテンプト ベー.フェー. Immune regulator
EP1138692A1 (en) 2000-03-29 2001-10-04 Erasmus Universiteit Rotterdam Fragments of human chorionic gonadotropin (hcg) as immunoregulator
US6844315B2 (en) 1998-05-20 2005-01-18 Erasmus Universiteit Rotterdam Immunoregulator
US7175679B2 (en) 2001-03-29 2007-02-13 Biotempt B.V. Oligopeptide treatment of NF-κB mediated inflammation
US20050227925A1 (en) 2004-04-08 2005-10-13 Robbert Benner Compositions capable of reducing elevated blood urea concentration
US6921751B1 (en) 1998-05-20 2005-07-26 Erasmus Universiteit Rotterdam Immunoregulator
US20030220258A1 (en) 2001-12-21 2003-11-27 Robbert Benner Treatment of ischemic events
US20090042807A1 (en) 1998-05-20 2009-02-12 Nisar Ahmed Khan Oligopeptide treatment of ischemia reperfusion injury
RU2139085C1 (en) 1998-06-23 1999-10-10 Санкт-Петербургская общественная организация "Институт биорегуляции и геронтологии" Agent stimulating reparative processes and method of its use
CA2689373A1 (en) 1998-09-17 2000-03-30 Hopital Sainte-Justine G protein-coupled receptor antagonists
AU1254700A (en) * 1998-11-20 2000-06-13 Mount Sinai Hospital Corporation Peptides that modulate the interaction of b class ephrins and pdz domains
US6507788B1 (en) * 1999-02-25 2003-01-14 Société de Conseils de Recherches et D'Applications Scientifiques (S.C.R.A.S.) Rational selection of putative peptides from identified nucleotide, or peptide sequences, of unknown function
CA2366678A1 (en) 1999-03-09 2000-09-14 Fornix Biosciences N.V. Synthetic complementary peptides and ophthalmologic uses thereof
US20030148955A1 (en) 1999-04-19 2003-08-07 Pluenneke John D. Soluble tumor necrosis factor receptor treatment of medical disorders
US6379970B1 (en) * 1999-04-30 2002-04-30 The Arizona Board Of Regents On Behalf Of The University Of Arizona Analysis of differential protein expression
RU2157233C1 (en) 1999-05-11 2000-10-10 Санкт-Петербургская общественная организация "Институт биорегуляции и геронтологии" Tetrapeptide showing geroprotective activity, pharmacological agent based on thereof and method of its use
US7994278B1 (en) 1999-08-06 2011-08-09 Nobel Biosciences Llc Biologically active polypeptides derived from a novel early stage pregnancy factor designated maternin (MA)
JP2003506411A (en) 1999-08-09 2003-02-18 トリペップ アクチ ボラゲット Protein polymerization inhibitors and methods of use
US6864355B1 (en) 2000-05-02 2005-03-08 Yale University Inhibition of NF-κB activation by blockade of IKKβ-NEMO interactions at the NEMO binding domain
EP1222199A4 (en) 1999-10-15 2003-03-12 Histatek Llc N-formyl peptide receptor complex with a g-protein kinase signal pathway modification agent
RU2155063C1 (en) 1999-10-20 2000-08-27 Санкт-Петербургская общественная организация "Институт биорегуляции и геронтологии" Tetrapeptide stimulating functional activity of neurons, pharmacological agent based on thereof and method of its use
DE19953339A1 (en) 1999-11-05 2001-05-17 Goldham Pharma Gmbh Secret fragments
AU781678C (en) 1999-11-18 2005-12-22 Ischemix, Inc. Compositions and methods for counteracting effects of reactive oxygen species and free radicals
DE60110396T2 (en) 2000-01-14 2006-01-19 Larson, Richard S., Albuquerque PEPTIDIC INHIBITORS OF LFA-1 / ICAM-1 INTERACTION
EP1263943A1 (en) 2000-02-11 2002-12-11 Eli Lilly & Company Protein c derivatives
AU2001245537A1 (en) 2000-03-10 2001-09-24 Monsanto Company Anti-hypertensive peptides
US7358330B2 (en) 2001-03-29 2008-04-15 Biotempt B.V. Immunoregulatory compositions
US20050037430A1 (en) 2000-03-29 2005-02-17 Biotempt B.V. Methods and uses for protein breakdown products
US7576174B2 (en) 2000-03-29 2009-08-18 Biotempt B.V. Compositions capable of reducing elevated blood urea concentration
US6783757B2 (en) 2000-06-01 2004-08-31 Kirkman Group, Inc. Composition and method for increasing exorphin catabolism to treat autism
US6586403B1 (en) 2000-07-20 2003-07-01 Salpep Biotechnology, Inc. Treating allergic reactions and inflammatory responses with tri-and dipeptides
US6539102B1 (en) * 2000-09-01 2003-03-25 Large Scale Proteomics Reference database
EP1217540A1 (en) * 2000-11-29 2002-06-26 Lafayette Software Inc. Methods of organizing data and processing queries in a database system, and database system and software product for implementing such method
US20020155106A1 (en) 2000-12-01 2002-10-24 Hammond David J. Method of identifying a ligand for a target molecule
US7316819B2 (en) 2001-03-08 2008-01-08 Unigene Laboratories, Inc. Oral peptide pharmaceutical dosage form and method of production
US6894028B2 (en) 2001-04-06 2005-05-17 Zengen, Inc. Use of KPV tripeptide for dermatological disorders
WO2002085117A1 (en) 2001-04-24 2002-10-31 Eisai Co., Ltd. Methods and compositions for preventing and treating septic shock and endotoxemia
US20030220259A1 (en) 2001-12-21 2003-11-27 Robbert Benner Treatment of neurological disorders
US7501391B2 (en) 2001-12-21 2009-03-10 Biotempt B.V. Treatment of transplant survival
US20030220257A1 (en) 2001-12-21 2003-11-27 Robbert Benner Treatment of trauma
US20030224995A1 (en) 2001-12-21 2003-12-04 Khan Nisar Ahmed Treatment of burns
US20030220260A1 (en) 2001-12-21 2003-11-27 Khan Nisar Ahmed Peptide compositions
US20080242837A1 (en) 2001-12-21 2008-10-02 Khan Nisar A Peptide compositions
US20080242618A1 (en) 2001-12-21 2008-10-02 Khan Nisar A Stratification
US20080194489A1 (en) 2001-12-21 2008-08-14 Khan Nisar A Treatment of iatrogenic disease
US7560433B2 (en) 2001-12-21 2009-07-14 Biotempt B.V. Treatment of multiple sclerosis (MS)
US20030220261A1 (en) 2001-12-21 2003-11-27 Khan Nisar Ahmed Treatment of iatrogenic disease
US20040013661A1 (en) 2001-12-21 2004-01-22 Gert Wensvoort Stratification
US7135286B2 (en) 2002-03-26 2006-11-14 Perlegen Sciences, Inc. Pharmaceutical and diagnostic business systems and methods
EP1494695A4 (en) 2002-04-15 2006-01-25 American Nat Red Cross Plasma protein-binding ligands
US20060173162A1 (en) 2002-08-15 2006-08-03 Rene Djurup Bactericidak anti-apoptotic, pro-inflammatory and anti-inflammatory peptides of heparin-binding protein (hbp)
US7517529B2 (en) 2003-04-08 2009-04-14 Biotempt B.V. Treatment of type I diabetes
IL157772A (en) 2003-09-04 2016-06-30 Bmr Solutions Ltd Compositions comprising an oligopeptide derived from tortoise spleen for use in stimulating mammalian hemopoiesis
US20090227505A1 (en) 2004-01-07 2009-09-10 Biotempt B.V. Methods and uses for protein breakdown products
SG163567A1 (en) 2005-07-05 2010-08-30 Biotempt Bv Treatment of tumors
PL2120991T3 (en) 2007-02-12 2014-07-31 Biotempt Bv Treatment of trauma hemorrhage with short oligopeptides

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3855397A (en) * 1973-04-12 1974-12-17 Allied Chem Method of producing sodium carbonate and bicarbonate spherules from brine
US3898328A (en) * 1973-12-03 1975-08-05 Syntex Inc Dry stable composition for the treatment of scours and dehydration
US3960830A (en) * 1973-12-06 1976-06-01 Hoechst Aktiengesellschaft Polyalkylene glycols used for the preparation of peptides
US4003989A (en) * 1974-05-06 1977-01-18 Bar On Ernest Pharmaceutical preparation
US4083951A (en) * 1976-05-05 1978-04-11 Beecham Group Limited Pharmaceutical composition
US4140760A (en) * 1976-11-09 1979-02-20 Reckitt & Colman Products Limited Pharmaceutical compositions for use in the suppression of gastric reflux
US4108846A (en) * 1977-02-01 1978-08-22 Hoffmann-La Roche Inc. Solid phase synthesis with base N alpha-protecting group cleavage
US4289750A (en) * 1978-10-16 1981-09-15 Kopp Klaus F Therapy of conditions which may be associated with altered renal function and dosage forms therefor
US4265874A (en) * 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4351762A (en) * 1981-03-10 1982-09-28 Bioresearch, Inc. Rapid, quantitative peptide synthesis using mixed anhydrides
US5137669A (en) * 1988-03-02 1992-08-11 Endocon, Inc. Manufacture of partially fused peptide pellet
US5610272A (en) * 1988-05-10 1997-03-11 Alpha-1 Biomedicals, Inc. Solid phase process for synthesizing thymosin α1
US5175254A (en) * 1988-09-24 1992-12-29 Societe D'expansion Scientifque Expansia Solid phase peptide synthesis using a polyacrylic support in aqueous solution
US5639480A (en) * 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5070187A (en) * 1989-11-03 1991-12-03 Trustees Of Boston University Pharmacologically effective antagonists of arginine-vasopressin
US5554378A (en) * 1990-09-03 1996-09-10 Takeda Chemical Industries, Ltd. Pharmaceutical composition and its mucosal use
US5200507A (en) * 1991-04-12 1993-04-06 Mallinckrodt Specialty Chemicals Company Method of separating a peptide from a resin
US5668111A (en) * 1992-12-07 1997-09-16 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5616608A (en) * 1993-07-29 1997-04-01 The United States Of America As Represented By The Department Of Health And Human Services Method of treating atherosclerosis or restenosis using microtubule stabilizing agent
US5665394A (en) * 1994-02-21 1997-09-09 Takeda Chemical Industries, Ltd. Matrix for sustained-release preparation
US5595760A (en) * 1994-09-02 1997-01-21 Delab Sustained release of peptides from pharmaceutical compositions
US5858375A (en) * 1994-09-15 1999-01-12 Medeva Europe Limited Pharmaceutical compositions containing D2 O
US5837218A (en) * 1995-09-15 1998-11-17 Resolution Pharmaceuticals Inc. Non-receptor cell mediated imaging agents
US20080267967A1 (en) * 2001-05-24 2008-10-30 Gorczynski Reginald M Modulation of cd200 receptors
US20030044463A1 (en) * 2001-09-06 2003-03-06 Romano Deghenghi Sustained release of microcrystalline peptide suspensions
US20110105415A1 (en) * 2001-10-04 2011-05-05 Biotempt B.V. Gene regulator
US7786084B2 (en) * 2001-12-21 2010-08-31 Biotempt B.V. Treatment of burns
US20100297258A1 (en) * 2001-12-21 2010-11-25 Biotempt B.V. Treatment of burns
US20030229022A1 (en) * 2002-06-07 2003-12-11 Fortuna Haviv Pharmaceutical formulation
US20090093398A1 (en) * 2003-11-07 2009-04-09 Jacques Bollekens Use of Fibroblast Growth Factor Fragments
US7795226B2 (en) * 2006-03-07 2010-09-14 Biotempt B.V. Control of radiation injury
US20110009344A1 (en) * 2006-03-07 2011-01-13 Biotempt B.V. Control of radiation injury
US20110113053A1 (en) * 2008-04-09 2011-05-12 Nisar Ahmed Khan Methods for identifying biologically active peptides and predicting their function

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE43309E1 (en) 2000-03-29 2012-04-10 Biotempt B.V. Immunoregulatory compositions
US8216998B2 (en) 2001-12-21 2012-07-10 Biotempt B.V. Treatment of ischemic events
US20070054860A1 (en) * 2001-12-21 2007-03-08 Biotempt B.V. Treatment of ischemic events
US20100004172A1 (en) * 2003-04-08 2010-01-07 Khan Nisar A Compositions for mucosal and oral administration comprising hcg fragments
US8540988B2 (en) 2006-02-13 2013-09-24 Alethia Biotherapeutics Inc. Antibodies that bind polypeptides involved in the process of bone remodeling
US9040246B2 (en) 2006-02-13 2015-05-26 Alethia Biotherapeutics Inc. Methods of making antibodies that bind polypeptides involved in the process of bone remodeling
US9695419B2 (en) 2006-02-13 2017-07-04 Daiichi Sankyo Company, Limited Polynucleotides and polypeptide sequences involved in the process of bone remodeling
US8431126B2 (en) 2006-02-13 2013-04-30 Alethia Biotherapeutics Inc. Antibodies that bind polypeptides involved in the process of bone remodeling
US7989160B2 (en) 2006-02-13 2011-08-02 Alethia Biotherapeutics Inc. Polynucleotides and polypeptide sequences involved in the process of bone remodeling
US9067984B2 (en) 2006-02-13 2015-06-30 Alethia Biotherapeutics Inc. Methods of impairing osteoclast differentiation using antibodies that bind Siglec-15
US8168181B2 (en) 2006-02-13 2012-05-01 Alethia Biotherapeutics, Inc. Methods of impairing osteoclast differentiation using antibodies that bind siglec-15
US8288341B2 (en) 2006-03-07 2012-10-16 Biotempt B.V. Control of radiation injury
US8900579B2 (en) 2009-10-06 2014-12-02 Alethia Biotherapuetics Inc. Siglec-15 antibodies in treating bone loss-related disease
US8741289B2 (en) 2009-10-06 2014-06-03 Alethia Biotherapeutics Inc. Siglec 15 antibodies in treating bone loss-related disease
US9388242B2 (en) 2009-10-06 2016-07-12 Alethia Biotherapeutics Inc. Nucleic acids encoding anti-Siglec-15 antibodies
US9617337B2 (en) 2009-10-06 2017-04-11 Daiichi Sankyo Company, Limited Siglec-15 antibodies in treating bone loss-related disease
USRE47672E1 (en) 2009-10-06 2019-10-29 Daiichi Sankyo Company, Limited Methods of impairing osteoclast differentiation using antibodies that bind siglec-15
US9493562B2 (en) 2012-07-19 2016-11-15 Alethia Biotherapeutics Inc. Anti-Siglec-15 antibodies

Also Published As

Publication number Publication date
US7358330B2 (en) 2008-04-15
USRE43309E1 (en) 2012-04-10
US20040208885A1 (en) 2004-10-21
US20080076719A1 (en) 2008-03-27

Similar Documents

Publication Publication Date Title
USRE43309E1 (en) Immunoregulatory compositions
EP1755634B1 (en) Oligopeptides for reducing elevated blood urea concentration
US8680059B2 (en) Oligopeptide acetate and formulations thereof
US7662776B2 (en) Treatment of tumors using short peptides from human chorionic gonadotropin (HCG)
CN102718858B (en) Glucagon-like peptide-1 (GLP-1) analogue monomer and dimer, preparation method therefor and application thereof
US7576174B2 (en) Compositions capable of reducing elevated blood urea concentration
NZ202757A (en) Peptides and medicaments
EP0347931B1 (en) Polypeptide comprising repeated cell-adhesive core sequences
JPH06228199A (en) Peptide binding body capable of passing through blood brain barrier
US5236903A (en) Polypeptide comprising repeated cell-adhesive core sequences
EP2058328B1 (en) Biologically active peptide VAPEEHPTLLTEAPLNPK derivatives
JP7046990B2 (en) Prodrug peptide with improved medicinal properties
KR20210093958A (en) Analogs targeting mitochondrial diseases
JP3002213B2 (en) Peptides and drugs containing this peptide
KR20070087267A (en) Biologically active peptide comprising tyrosyl-seryl-valine (ysv)
USRE43279E1 (en) Compositions capable of reducing elevated blood urea concentration
WO2022042590A1 (en) Polypeptides for repairing skin wounds or mucosal injuries, and applications thereof
CN114989259A (en) Small molecular peptide Ped4 and application thereof
JPH06172203A (en) Agent for inhibiting abnormal cell producing fibronectin receptor
JP2777647B2 (en) Cancer metastasis inhibitor
JPH083190A (en) Polypeptide composed of recurring structure of cell-adhesive core sequence

Legal Events

Date Code Title Description
AS Assignment

Owner name: ERASMUS UNIVERSITEIT ROTTERDAM, NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KHAN, NISAR AHMED;BENNER, ROBBERT;REEL/FRAME:020569/0391

Effective date: 20040604

AS Assignment

Owner name: BIOTEMPT B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ROTTERDAM, ERASMUS UNIVERSITEIT;REEL/FRAME:020677/0987

Effective date: 20071018

STCB Information on status: application discontinuation

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