WO2006062685A2 - Novel peptides that bind to the erythropoietin receptor - Google Patents

Novel peptides that bind to the erythropoietin receptor Download PDF

Info

Publication number
WO2006062685A2
WO2006062685A2 PCT/US2005/041112 US2005041112W WO2006062685A2 WO 2006062685 A2 WO2006062685 A2 WO 2006062685A2 US 2005041112 W US2005041112 W US 2005041112W WO 2006062685 A2 WO2006062685 A2 WO 2006062685A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
peg
epo
molecular weight
daltons
Prior art date
Application number
PCT/US2005/041112
Other languages
French (fr)
Other versions
WO2006062685A3 (en
Inventor
Christopher P. Holmes
Qun Yin
Genet Zemede
Ashok Bhandari
Yaohua S. Dong
David Tumelty
Guy Lalonde
Balu Palani
Peter J. Schatz
Nicholas C. Wrighton
William J. Dower
Brian T. Frederick
Anjan Chakrabarti
Original Assignee
Affymax, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Affymax, Inc. filed Critical Affymax, Inc.
Publication of WO2006062685A2 publication Critical patent/WO2006062685A2/en
Publication of WO2006062685A3 publication Critical patent/WO2006062685A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to peptide compounds that are agonists of the erythropoietin receptor (EPO-R).
  • EPO-R erythropoietin receptor
  • the invention further relates to therapeutic methods using such peptide compounds to treat disorders associated with insufficient or defective red blood cell production.
  • Pharmaceutical compositions, which comprise the peptide compounds of the invention, are also provided.
  • EPO Erythropoietin
  • kD kilodaltons
  • ⁇ , ⁇ , and asialo
  • the ⁇ and ⁇ forms differ slightly m their carbohydrate components, but have the same potency, biological activity, and molecular weight
  • the asialo form is an ⁇ or ⁇ form with the terminal carbohydrate (sialic acid) removed.
  • the DNA sequences encoding EPO have been reported [U S. Pat. No. 4,703,008 to Lm]
  • EPO stimulates mitotic division and differentiation of erythrocyte precursor cells, and thus ensures the production of erythrocytes It is produced in the kidney when hypoxic conditions prevail.
  • EPO-mduced differentiation of erythrocyte precursor cells globm synthesis is induced; heme complex synthesis is stimulated; and the number of ferritin receptors increases.
  • These changes allow the cell to take on more iron and synthesize functional hemoglobin, which binds in mature erythrocytes oxygen.
  • erythrocytes and their hemoglobin play a key role in supplying the body with oxygen.
  • EPO is present in very low concentrations in plasma when the body is in a healthy state, in which tissues receive sufficient oxygenation from the existing number of erythrocytes. This normal low EPO concentration is sufficient to stimulate replacement of red blood cells that are normally lost through aging.
  • the amount of EPO m the circulation is increased under conditions of hypoxia when oxygen transport by blood cells in circulation is reduced Hypoxia may be caused, for example, by substantial blood loss through hemorrhage, destruction of red blood cells by over-exposure to radiation, reduction in oxygen intake due to high altitude or prolonged unconsciousness, or vanous forms of anemia.
  • hypoxia may be caused, for example, by substantial blood loss through hemorrhage, destruction of red blood cells by over-exposure to radiation, reduction in oxygen intake due to high altitude or prolonged unconsciousness, or vanous forms of anemia.
  • elevated EPO levels increase red blood cell production by stimulating the proliferation of erythroid progenitor cells.
  • EPO levels in circulation are decreased.
  • EPO is essential in the process of red blood cell formation, this hormone has potentially useful applications in both the diagnosis and treatment of blood disorders characterized by low or defective red blood cell production.
  • Recent studies have provided a basis for the projection of EPO therapy efficacy for a variety of disease states, disorders, and states of hematologic irregularity, including: beta-thalassemia [See Vedovato, et al. (1984) Acta. Haematol. 71:21 1-213]; cystic fibrosis [See Vichinsky, et al. (1984) J. Pediatric 105:15-21]; pregnancy and menstrual disorders [See Cotes, el al. (193) B ⁇ t. J. Ostet.
  • EPO The biological effect of EPO appears to be mediated, in part, by interaction with a cell membrane bound receptor.
  • Initial studies using immature erythroid cells isolated from mouse spleen suggest that the EPO-bmding cell surface proteins comprise two polypeptides having approximate molecular weights of 85,000 Daltons and 100,000 Daltons, respectively [Sawyer, et al. (1987) Proc. Natl. Acad. Sci. USA 84:3690-3694].
  • the number of EPO binding sites was calculated to average from 800 to 1000 per cell surface Of these binding sites, approximately 300 bound EPO with a K d value of approximately 90 picomolar (pM), while the remaining sites bound EPO with a reduced affinity of approximately 570 pM [Sawyer, et al.
  • EPO-R EPO receptor
  • peptides containing a peptide motif have been identified, members of which bind to EPO-R and stimulate EPO-dependent cell proliferation. Yet, peptides identified to date as containing the motif stimulate EPO-dependent cell proliferation in vitro with EC50 values between about 20 nanomolar (nM) and 25OnM. Thus, peptide concentrations of 2OnM to 25OnM are required to stimulate 50% of the maximal cell proliferation stimulated by EPO Given the immense potential of EPO-R agonists, both for studies of the important biological activities mediated by this receptor and for treatment of disease, there remains a need for the identification of peptide EPO-R agonists of enhanced potency and activity. The present invention provides such compounds.
  • the present invention provides EPO-R agonist monome ⁇ c peptides of dramatically enhanced potency and activity and dime ⁇ c peptide agonists that comprise two peptide monomers.
  • the potency of these novel peptide agonists may be further enhanced by one or more modifications, including: acetylation, intramolecular disulfide bond formation, covalent attachment of one or more polyethylene glycol (PEG) moieties, and others as listed in Figures IA - IL and throughout this application.
  • the invention also provides peptides with protecting groups and/or hydrophobic groups. Protecting groups and/or hydrophobic groups associated with the peptides can be used to prolong half-lives of the peptides in circulation, and facilitate uptake by cells and transport across cell membranes.
  • the invention further provides pharmaceutical compositions comprised of such peptide agonists, and methods to treat various medical conditions using such peptide agonists.
  • Figures IA - IL show a table of peptides, including peptide sequences of the present invention. Peptide sequences are provided using the single-letter amino acid code. Modified and non-naturally occu ⁇ ng ammo acids are indicated using the abbreviations defined, infra, in this specification. For convenience, each individual peptide is referred to by reference to its unique sequence identification number (SEQ ID NO) given in the far left-hand column.
  • SEQ ID NO unique sequence identification number
  • Linker moieties of the individual peptides when present, are specified in the column labeled "Linker.”
  • the column labeled “Lmker-R” indicates the chemical moiety present as the R group, if present, on the linker.
  • polypeptide or "protein” refers to a polymer of ammo acid monomers that are alpha ammo acids joined together through amide bonds. Polypeptides are therefore at least two amino acid residues in length, and are usually longer. Generally, the term “peptide” refers to a polypeptide that is only a few amino acid residues in length.
  • the novel EPO-R agonist peptides of the present invention are preferably no more than about 50 amino acid residues in length. They are more preferably from about 8 to about 45 amino acid residues in length.
  • a polypeptide, in contrast with a peptide, may comprise any number of amino acid residues. Hence, the term polypeptide included peptides as well as longer sequences of amino acids.
  • the phrase “pharmaceutically acceptable” refers to molecular entities and compositions that are "generally regarded as safe,” e g , that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a human.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U S Pharmacopeia or other generally recognized pharmacopeia for use m animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin
  • agonist refers to a biologically active hgand which binds to its complementary biologically active receptor and activates the latter either to cause a biological response in the receptor, or to enhance preexisting biological activity of the receptor.
  • Novel peptides that are EPO-R agonists
  • the present invention relates to peptides that are agonists of the EPO-R and show dramatically enhanced potency and activity.
  • These peptide agonists are preferably of about 8 to about 45 amino acids in length.
  • the peptides of this invention may be monomers, homo- or hetero- dimers, or other homo- or hetero- multimers.
  • the term "homo” means comprising identical monomers; thus, for example, a homodimer of the present invention is a peptide comprising two identical monomers.
  • the term “hetero” means comprising different monomers; thus, for example, a heterodimer of the present invention is a peptide comprising two non-identical monomers.
  • the peptide multimers of the invention may be trimers, tetramers, pentamers, or other higher order structures. Moreover, such dimers and other multimers may be heterodimers or heteromultimers.
  • the peptide monomers of the present invention may be degradation products (e.g., oxidation products of methionine or deamidated glutamine, arganine, and C-terminus amide). Such degradation products may be used in and are therefore considered part of the present invention.
  • the heteromultimers of the invention comprise multiple peptides that are all EPO-R agonist peptides.
  • the multimers of the invention are homomultimers: i.e., they comprise multiple EPO-R agonist peptides of the same amino acid sequence. Accordingly, the present invention also relates to homo- or hetero- dimeric peptide agonists of
  • the dimers of the invention comprise two peptides that are both EPO-R agonist peptides. These preferred dimeric peptide agonists comprise two peptide monomers, wherein each peptide monomer is of about 8 to about 45 amino acids in length. In particularly preferred embodiments, the dimers of the invention comprise two EPO-R agonist peptides of the same amino acid sequence.
  • Stereoisomers e.g., D-amino acids of the twenty conventional amino acids, unnatural amino acids such as a,a-disubstituted amino acids, N-alkyl amino acids, lactic acid, and other unconventional amino acids may also be suitable components for compounds of the present invention.
  • unconventional amino acids include, but are not limited to: ⁇ -alanine, 3-py ⁇ dylalanine, A- hydroxyprolme, 0-phosphose ⁇ ne, N-methylglycine, N-acetylse ⁇ ne, N-formylmethionine, 3- methylhistidine, 5-hydroxylysine, nor-leucine, and other similar amino acids and imino acids.
  • a preferred amino terminal modification is acetylation (e g , with acetic acid or a halogen substituted acetic acid).
  • an N-terminal glycine is acetylated to N-acetylglycine (AcG).
  • a the C-terminal glycine is N-methylglycme (MeG, also known as sarcosine).
  • the peptide monomers of the invention contain an intramolecular disulfide bond between the two cysteine residues of the core sequence.
  • the present invention also provides conjugates of these peptide monomers
  • the monome ⁇ c peptides of the present invention are dime ⁇ zed or ohgome ⁇ zed, thereby enhancing EPO-R agonist activity.
  • the peptide monomers of the invention may be oligome ⁇ zed using the biotin/streptavidin system Biotinylated analogs of peptide monomers may be synthesized by standard techniques.
  • the peptide monomers may be C-terminally biotinylated.
  • biotinylated monomers are then ohgome ⁇ zed by incubation with streptavidin [e g , at a 4' 1 molar ratio at room temperature in phosphate buffered saline (PBS) or HEPES-buffered RPMI medium (Invitrogen) for 1 hour]
  • streptavidin e g , at a 4' 1 molar ratio at room temperature in phosphate buffered saline (PBS) or HEPES-buffered RPMI medium (Invitrogen) for 1 hour
  • biotinylated peptide monomers may be oligome ⁇ zed by incubation with any one of a number of commercially available anti-biotin antibodies [e g , goat anti- biotin IgG from Kirkegaard & Perry Laboratories, Inc (Washington, DC)] .
  • the peptide monomers of the invention are dime ⁇ zed by covalent attachment to at least one linker moiety.
  • the linker (L ⁇ ) moiety is preferably, although not necessarily, a C M2 linking moiety optionally terminated with one or two -NH- linkages and optionally substituted at one or more available carbon atoms with a lower alkyl substituent.
  • the linker L ⁇ comprises -NH-R-NH- wherein R is a lower (Ci -6 ) alkylene substituted with a functional group such as a carboxyl group or an amino group that enables binding to another molecular moiety (e g , as may be present on the surface of a solid support).
  • the linker is a lysine residue or a lysine amide (a lysine residue wherein the carboxyl group has been converted to an amide moiety - CONH 2 ).
  • the linker bridges the C-termini of two peptide monomers, by simultaneous attachment to the C-termmal amino acid of each monomer.
  • the dimer when the C-terminal linker L ⁇ is a lysine amide the dimer may be illustrated structurally as shown in Formula I, and summarized as shown in Formula II: Formula I Formula II
  • N 2 represents the nitrogen atom of lysine's ⁇ -ammo group and N 1 represents the nitrogen atom of lysine's ⁇ -amino group.
  • the dimeric structure can be written as [peptide] 2 Lys-amide to denote a peptide bound to both the ⁇ and ⁇ amino groups of lysine, or [Ac- peptide] 2 Lys-amide to denote an N-terminally acetylated peptide bound to both the ⁇ and ⁇ amino groups of lysine, or [Ac-peptide, disulfide] 2 Lys-amide to denote an N-termmally acetylated peptide bound to both the a and ⁇ ammo groups of lysine with each peptide containing an intramolecular disulfide loop, or [Ac-peptide, disulfide] 2 Lys-spacer-PEG to denote an N-terminally acetylated peptide bound to both
  • polyethylene glycol may serve as the linker L ⁇ that dime ⁇ zes two peptide monomers, for example, a single PEG moiety may be simultaneously attached to the ⁇ -termini of both peptide chains of a peptide dimer.
  • the linker (L ⁇ ) moiety is preferably, but not necessarily, a molecule containing two carboxyhc acids and optionally substituted at one or more available atoms with an additional functional group such as an amine capable of being bound to one or more PEG molecules. Such a molecule can be depicted as.
  • n is an integer from 0 to 10
  • m is an integer from 1 to 10
  • X is selected from O, S, ⁇ (CH 2 ) p ⁇ Ri, NCO(CH 2 ) P NR,, and CHNR 1 , R, is selected from H, Boc, Cbz, etc.
  • p is an integer from 1 to 10.
  • one amino group of each of the peptides form an amide bond with the linker L ⁇ .
  • the amino group of the peptide bound to the linker L ⁇ is the epsilon amine of a lysine residue or the alpha amine of the N-terminal residue, or an amino group of the optional spacer molecule.
  • both n and m are one, X is NCO(CH 2 ) p NRi , p is two, and Ri is Boc.
  • a dimeric EPO peptide containing such a preferred linker may be structurally illustrated as shown in Formula III.
  • the Boc group can be removed to liberate a reactive amine group capable of forming a covalent bond with a suitably activated water soluble polymer species, for example, a PEG species such as mPEG-para-mtrophenylcarbonate (mPEG-NPC), mPEG-succmimidyl propionate (mPEG- SPA), and N-hydroxysuccinimide-PEG (NHS-PEG) (see, e.g., US Patent No. 5,672,662).
  • mPEG-NPC mPEG-para-mtrophenylcarbonate
  • mPEG-SPA mPEG-succmimidyl propionate
  • NHS-PEG N-hydroxysuccinimide-PEG
  • a dimeric EPO peptide containing such a preferred linker may be structurally illustrated as shown in Formula IV.
  • peptide dimers will also contain one or more intramolecular disulfide bonds between cysteine residues of the peptide monomers.
  • the two monomers contain at least one intramolecular disulfide bond.
  • both monomers of a peptide dimer contain an intramolecular disulfide bond, such that each monomer contains a cyclic group
  • a peptide monomer or dimer may further comprise one or more spacer moieties.
  • spacer moieties may be attached to a peptide monomer or to a peptide dimer.
  • spacer moieties are attached to the linker L ⁇ moiety that connects the monomers of a peptide dimer.
  • spacer moieties may be attached to a peptide dimer via the carbonyl carbon of a lysine linker, or via the nitrogen atom of an iminodiacetic acid linker.
  • a spacer may connect the linker of a peptide dimer to an attached water soluble polymer moiety or a protecting group.
  • such a spacer may connect a peptide monomer to an attached water soluble polymer moiety.
  • the spacer moiety is a Ci. 12 linking moiety optionally terminated with - NH- linkages or carboxyl (-COOH) groups, and optionally substituted at one or more available carbon atoms with a lower alkyl substituent.
  • the spacer is R-COOH wherein R is a lower (C 1 - I5 ) alkylene optionally substituted with a functional group such as a carboxyl group or an amino group that enables binding to another molecular moiety.
  • the spacer may be a glycine (G) residue, or an amino hexanoic acid.
  • the amino hexanoic acid is 6-amino hexanoic acid (Ahx)
  • the spacer 6-amino hexanoic acid (Ahx) is bound to the N- terminus of a peptide
  • the peptide terminal amine group may be linked to the carboxyl group of Ahx via a standard amide coupling.
  • the amine of Ahx may be linked to the carboxyl group of the linker via a standard amide coupling.
  • the structure of such a peptide may be depicted as shown in Formula V, and summarized as shown in Formula VI.
  • the spacer is -NH-R-NH- wherein R is a lower (Q _ 6 ) alkylene substituted with a functional group such as a carboxyl group or an ammo group that enables binding to another molecular moiety.
  • R is a lower (Q _ 6 ) alkylene substituted with a functional group such as a carboxyl group or an ammo group that enables binding to another molecular moiety.
  • the spacer may be a lysine (K) residue or a lysine amide
  • the spacer moiety has the following structure:
  • ⁇ , ⁇ , ⁇ , and ⁇ are each integers whose values are independently selected.
  • ⁇ , ⁇ , and ⁇ are each integers whose values are independently selected from one to about six, ⁇ is zero or one, ⁇ is an integer selected from zero to about ten, except that when ⁇ is greater than one, ⁇ is two, and Y is selected from NH or CO.
  • ⁇ , ⁇ , and ⁇ are each equal to two, both ⁇ and ⁇ are equal to 1 , and Y is NH.
  • a peptide dimer containing such a spacer is illustrated schematically in Formula VII, where the linker is a lysine and the spacer joins the linker to a Boc protecting group.
  • ⁇ and ⁇ are zero, ⁇ and ⁇ together equal five, and Y is CO.
  • the linker plus spacer moiety has the structure shown in Formula VIII or Formula IX Formula VIII Formula IX
  • the peptide monomers, dimers, or multimers of the invention may further comprise one or more water soluble polymer moieties.
  • these polymers are covalently attached to the peptide compounds of the invention
  • the polymer will be pharmaceutically acceptable.
  • One skilled m the art will be able to select the desired polymer based on such considerations as whether the polymer-peptide conjugate will be used therapeutically, and if so, the desired dosage, circulation time, resistance to proteolysis, and other considerations.
  • the water soluble polymer may be, for example, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-l,3-dioxolane, poly-l,3,6-t ⁇ oxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), poly(n-vinyl pyrrohdone)polyethylene glycol, propropylene glycol homopolymers, polypropylene oxide/ethylene oxide copolymers, and polyoxyethylated polyols.
  • PEG polyethylene glycol
  • copolymers of ethylene glycol/propylene glycol carboxymethylcellulose
  • dextran polyvinyl alcohol
  • polyvinyl pyrrolidone poly-l,3-dioxolane
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • a preferred PEG for use m the present invention comprises linear, unbranched PEG having a molecular weight that is greater than 10 kilodaltons (kD) and is more preferably between about 20 and 60 kD in molecular weight. Still more preferably, the linear unbranched PEG moiety should have a molecular weight of between about 20 and 40 kD, with 20 kD PEG being particularly preferred. It is understood that in a given preparation of PEG, the molecular weights will typically vary among individual molecules. Some molecules will weight more, and some less, than the stated molecular weight. Such variation is generally reflect by use of the word "about” to describe molecular weights of the PEG molecules.
  • the number of polymer molecules attached may vary; for example, one, two, three, or more water soluble polymers may be attached to an EPO-R agonist peptide of the invention.
  • the multiple attached polymers may be the same or different chemical moieties (e g , PEGs of different molecular weight)
  • the invention contemplates EPO-R agonist peptides having two or more PEG moieities attached thereto
  • both of the PEG moietieis are linear, unbranched PEG each preferably having a molecular weight of between about 10 and about 60 kD.
  • each linear unbranched PEG moiety has a molecular weight that is between about 20 and 40 kD, and still more preferably between about 20 and 30 kD with a molecular weight of about 20 kD for each linear PEG moiety being particularly preferred.
  • molecular weights for PEG are also contemplated in such embodiments.
  • the invention contemplates and encompasses EPO-R agonist peptides having two or more linear unbranched PEG moieties attached thereto, at least one or both of which has a molecular weight between about 20 and 40 kD or between about 20 and 30 kD
  • the invention contemplates and encompasses EPO-R agonist peptides having two or more linear unbranched PEG moieties attached thereto, at least one of which has a molecular weight between about 40 and 60 kD.
  • PEG may serve as a linker that dime ⁇ zes two peptide monomers.
  • PEG is attached to at least one terminus (N-termmus or C-terminus) of a peptide monomer or dimer.
  • PEG is attached to a spacer moiety of a peptide monomer or dimer.
  • PEG is attached to the linker moiety of a peptide dimer.
  • PEG is attached to a spacer moiety, where said spacer moiety is attached to the linker L ⁇ moiety that connects the monomers of a peptide dimer.
  • PEG is attached to a spacer moiety, where said spacer moiety is attached to a peptide dimer via the carbonyl carbon of a lysine linker, or the amide nitrogen of a lysine amide linker.
  • the peptide sequences of the present invention can be present alone or in conjunction with N- termmal and/or C-termmal extensions of the peptide chain.
  • Such extensions may be naturally encoded peptide sequences optionally with or substantially without non-naturally occurring sequences; the extensions may include any additions, deletions, point mutations, or other sequence modifications or combinations as desired by those skilled in the art.
  • naturally- occurring sequences may be full-length or partial length and may include amino acid substitutions to provide a site for attachment of carbohydrate, PEG, other polymer, or the like via side chain conjugation
  • the amino acid substitution results in humanization of a sequence to make in compatible with the human immune system.
  • Fusion proteins of all types are provided, including immunoglobulin sequences adjacent to or in near proximity to the EPO-R activating sequences of the present invention with or without a non-immunoglobulm spacer sequence.
  • One type of embodiment is an immunoglobulin chain having the EPO-R activating sequence in place of the variable (V) region of the heavy and/or light chain.
  • the peptides of the invention may be prepared by classical methods known in the art. These standard methods include exclusive solid phase synthesis, partial solid phase synthesis methods, fragment condensation, classical solution synthesis, and recombinant DNA technology [See, e g , Mer ⁇ field J Am Chem Soc 1963 85-2149]
  • the peptide monomers of a peptide dimer are synthesized individually and dime ⁇ zed subsequent to synthesis. In preferred embodiments the peptide monomers of a dimer have the same ammo acid sequence
  • the peptide monomers of a dimer are linked via their C-termini by a linker L ⁇ moiety having two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety (e g , as may be present on the surface of a solid support).
  • the two peptide monomers may be synthesized directly onto two reactive nitrogen groups of the linker L ⁇ moiety in a variation of the solid phase synthesis technique. Such synthesis may be sequential or simultaneous.
  • the protected diamine is a protected lysine.
  • the protected linker is coupled to a solid support via the linker's third functional group
  • the first amine protecting group is removed, and the first peptide of the dimer is synthesized on the first deprotected amine moiety.
  • the second amine protecting group is removed, and the second peptide of the dimer is synthesized on the second deprotected amine moiety.
  • the first amino moiety of the linker may be protected with Alloc, and the second with Fmoc.
  • the Fmoc group (but not the Alloc group) may be removed by treatment with a mild base [e g , 20% pipe ⁇ dine in dimethyl formamide (DMF)], and the first peptide chain synthesized Thereafter the Alloc group may be removed with a suitable reagent [e g , Pd(PPh 3 )/4-methyl morpholme and chloroform], and the second peptide chain synthesized
  • a mild base e g , 20% pipe ⁇ dine in dimethyl formamide (DMF)
  • DMF dimethyl formamide
  • a suitable reagent e g , Pd(PPh 3 )/4-methyl morpholme and chloroform
  • two amine functional groups of the linker molecule are protected with the same removable amine protecting group
  • the protected diamine is a protected lysine.
  • the protected linker is coupled to a solid support via the linker's third functional group.
  • the two protected functional groups of the linker molecule are simultaneously deprotected, and the two peptide chains simultaneously synthesized on the deprotected amines. Note that using this technique, the sequences of the peptide chains of the dimer will be identical, and the thiol-protectmg groups for the cysteine residues are all the same.
  • a preferred method for peptide synthesis is solid phase synthesis.
  • Solid phase peptide synthesis procedures are well-known in the art [see, e g , Stewart Solid Phase Peptide Syntheses (Freeman and Co.: San Francisco) 1969; 2002/2003 General Catalog from Novabiochem Corp, San Diego, USA; Goodman Synthesis of Peptides and Peptidomimetics (Houben-Weyl, Stuttgart) 2002].
  • synthesis is typically commenced from the C-termmal end of the peptide using an ⁇ -amino protected resin.
  • a suitable starting material can be prepared, for instance, by attaching the required ⁇ -amino acid to a chloromethylated resin, a hydroxymethyl resm, a polystyrene resin, a benzhydrylamine resin, or the like.
  • a chloromethylated resin is sold under the trade name BIO-BEADS SX-I by Bio Rad Laboratories (Richmond, CA).
  • BIO-BEADS SX-I Bio Rad Laboratories (Richmond, CA).
  • BIO-BEADS SX-I Bio Rad Laboratories (Richmond, CA).
  • the preparation of the hydroxymethyl resm has been described [Bodonszky, et al. (1966) Chem. Ind. London 38: 1597]
  • the benzhydrylamine (BHA) resin has been described [Pietta and Marshall (1970) Chem. Commun.
  • an ⁇ -ammo protected amino acid may be coupled to a chloromethylated resin with the aid of a cesium bicarbonate catalyst, according to the method described by Gisin (1973) HeIv. Chim Acta 56: 1467.
  • the ⁇ -amino protecting group is removed, for example, using trifluoroacetic acid (TFA) or hydrochloric acid (HCl) solutions in organic solvents at room temperature Thereafter, ⁇ -ammo protected amino acids are successively coupled to a growing support-bound peptide chain.
  • TFA trifluoroacetic acid
  • HCl hydrochloric acid
  • the ⁇ -amino protecting groups are those known to be useful in the art of stepwise synthesis of peptides, including" acyl-type protecting groups (e g , formyl, t ⁇ fluoroacetyl, acetyl), aromatic urethane-type protecting groups [e g , benzyloxycarboyl (Cbz) and substituted Cbz], aliphatic urethane protecting groups [e g , t-butyloxycarbonyl (Boc), isopropyloxycarbonyl, cyclohexyloxycarbonyl], and alkyl type protecting groups (e g , benzyl, t ⁇ phenylmethyl), fluorenylmethyl oxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), and l-(4,4-dimethyl-2,6- dioxocyclohex-l-yhdene)ethyl (Dde).
  • the side chain protecting groups (typically ethers, esters, t ⁇ tyl, PMC (2,2,5,7,8- pentamethyl-chroman-6-sulphonyl), and the like) remain intact during coupling and is not split off during the deprotection of the amino-terminus protecting group or during coupling.
  • the side chain protecting group must be removable upon the completion of the synthesis of the final peptide and under reaction conditions that will not alter the target peptide.
  • the side chain protecting groups for Tyr include tetrahydropyranyl, tert-butyl, trityl, benzyl, Cbz, Z-Br-Cbz, and 2,5-dichlorobenzyl.
  • the side chain protecting groups for Asp include benzyl, 2,6-dichlorobenzyl, methyl, ethyl, and cyclohexyl.
  • the side chain protecting groups for Thr and Ser include acetyl, benzoyl, trityl, tetrahydropyranyl, benzyl, 2,6-dichlorobenzyl, and Cbz
  • the side chain protecting groups for Arg include nitro, Tosyl (Tos), Cbz, adamantyloxycarbonyl mesitoylsulfonyl (Mts), 2,2,4,6,7- pentamethyldihydrobenzofurane-5-sulfonyl (Pbf), 4-mthoxy-2,3,6-trimethyl-benzenesulfonyl (Mtr), or Boc.
  • the side chain protecting groups for Lys include Cbz, 2-chlorobenzyloxycarbonyl (2-Cl-Cbz), 2-bromobenzyloxycarbonyl (2-Br-Cbz), Tos, or Boc After removal of the ⁇ -ammo protecting group, the remaining protected ammo acids are coupled stepwise in the desired order.
  • Each protected amino acid is generally reacted in about a 3-fold excess using an appropriate carboxyl group activator such as 2-(lH-benzotriazol-l-yl)-l , l ,3,3 tetramethyluronium hexafluorophosphate (HBTU) or dicyclohexylcarbodimide (DCC) in solution, for example, in methylene chloride (CH 2 Cl 2 ), N-methyl pyrrolidone, dimethyl formamide (DMF), or mixtures thereof.
  • an appropriate carboxyl group activator such as 2-(lH-benzotriazol-l-yl)-l , l ,3,3 tetramethyluronium hexafluorophosphate (HBTU) or dicyclohexylcarbodimide (DCC) in solution, for example, in methylene chloride (CH 2 Cl 2 ), N-methyl pyrrolidone, dimethyl formamide (DMF), or mixtures thereof.
  • the desired peptide is decoupled from the resin support by treatment with a reagent, such as t ⁇ fiuoroacetic acid (TFA) or hydrogen fluoride (HF), which not only cleaves the peptide from the resin, but also cleaves all remaining side chain protecting groups
  • a reagent such as t ⁇ fiuoroacetic acid (TFA) or hydrogen fluoride (HF)
  • FFA t ⁇ fiuoroacetic acid
  • HF hydrogen fluoride
  • the side chain protected peptide can be decoupled by treatment of the peptide resin with ammonia to give the desired side chain protected amide or with an alkylamine to give a side chain protected alkylamide or dialkylamide.
  • Synthetic ammo acids that can be substituted into the peptides of the present invention include, but are not limited to, N-methyl, L-hydroxypropyl, L-3, A- dihydroxyphenylalanyl, ⁇ amino acids such as L- ⁇ -hydroxylysyl and D- ⁇ -methylalanyl, L- ⁇ - methylalanyl, ⁇ amino acids, and isoquinolyl D-amino acids and non-naturally occurring synthetic amino acids can also be incorporated into the peptides of the present invention.
  • Ammo terminus modifications include methylation (e g , -NHCH 3 or ⁇ N(CH 3 ) 2 ), acetylation (e g , with acetic acid or a halogenated derivative thereof such as ⁇ -chloroacetic acid, ⁇ -bromoacetic acid, or ⁇ -iodoacetic acid), adding a benzyloxycarbonyl (Cbz) group, or blocking the ammo terminus with any blocking group containing a carboxylate functionality defined by RCOO-- or sulfonyl functionality defined by R-SO 2 --, where R is selected from alkyl, aryl, heteroaryl, alkyl aryl, and the like, and similar groups
  • Carboxy terminus modifications include replacing the free acid with a carboxamide group or forming a cyclic lactam at the carboxy terminus to introduce structural constraints
  • One can also cychze the peptides of the invention, or incorporate a desammo or descarboxy residue at the termini of the peptide, so that there is no terminal amino or carboxyl group, to decrease susceptibility to proteases or to restrict the conformation of the peptide C-terminal functional groups of the compounds of the present invention include amide, amide lower alkyl, amide di(lower alkyl), lower alkoxy, hydroxy, and carboxy, and the lower ester derivatives thereof, and the pharmaceutically acceptable salts thereof
  • proline analogues in which the ring size of the proline residue is changed from 5 members to 4, 6, or 7 members can be employed
  • Cyclic groups can be saturated or unsaturated, and if unsaturated, can be aromatic or non-aromatic Heterocyclic groups preferably contain one or more nitrogen, oxygen, and/or sulfur heteroatoms Examples of such groups include the furazanyl, furyl, lmidazohdinyl, lmidazolyl, lmidazohnyl, isothi
  • the peptide compounds of the invention also serve as structural models for non-peptidic compounds with similar biological activity
  • Those of skill in the art recognize that a variety of techniques are available for constructing compounds with the same or similar desired biological activity as the lead peptide compound, but with more favorable activity than the lead with respect to solubility, stability, and susceptibility to hydrolysis and proteolysis [See, Morgan and Gamor (1989) Ann Rep. Med Chem. 24:243-252]. These techniques include replacing the peptide backbone with a backbone composed of phosphonates, amidates, carbamates, sulfonamides, secondary amines, and N- methylamino acids.
  • the compounds of the present invention may contain one or more intramolecular disulfide bonds.
  • a peptide monomer or dimer comprises at least one intramolecular disulfide bond.
  • a peptide dimer comprises two intramolecular disulfide bonds.
  • Such disulfide bonds may be formed by oxidation of the cysteine residues of the peptide core sequence.
  • control of cysteine bond formation is exercised by choosing an oxidizing agent of the type and concentration effective to optimize formation of the desired isomer. For example, oxidation of a peptide dimer to form two intramolecular disulfide bonds (one on each peptide chain) is preferentially achieved (over formation of intermolecular disulfide bonds) when the oxidizing agent is DMSO.
  • the formation of cysteine bonds is controlled by the selective use of thiol-protecting groups during peptide synthesis.
  • the first monomer peptide chain is synthesized with the two cysteine residues of the core sequence protected with a first thiol protecting group [e g , trityl(Trt), allyloxycarbonyl (Alloc), and l-(4,4-dimethyl-2,6-dioxocyclohex-l-yhdene)ethyl (Dde) or the like]
  • the second monomer peptide is synthesized the two cysteine residues of the core sequence protected with a second thiol protecting group different from the first thiol protecting group [e g , acetamidomethyl (Acm), t-butyl (tBu), or the like].
  • the first thiol protecting groups are removed effecting bis
  • analogues of these disulfide derivatives in which one of the sulfurs has been replaced by a CH 2 group or other isotere for sulfur.
  • These analogues can be prepared from the compounds of the present invention, wherein each core sequence contains at least one C or homocysteine residue and an ⁇ -amino- ⁇ -buty ⁇ c acid in place of the second C residue, via an intramolecular or intermolecular displacement, using methods known m the art [See, e g , Barker, et al. (1992) J. Med. Chem. 35:2040-2048 and Or, et al. (1991) J. Org. Chem.
  • a peptide may be synthesized wherein one cysteine of the core sequence is replaced with lysine and the second cysteine is replaced with glutamic acid Thereafter a cyclic monomer may be formed through an amide bond between the side chains of these two residues
  • a peptide may be synthesized wherein one cysteine of the core sequence is replaced with lysine.
  • a cyclic monomer may then be formed through a thio-ether linkage between the side chains of the lysine residue and the second cysteine residue of the core sequence
  • amide-cychzation strategies and thio-ether cychzation strategies can both be readily used to cyclize the compounds of the present invention.
  • the amino-terminus of the peptide can be capped with an ⁇ -substituted acetic acid, wherein the ⁇ -substituent is a leaving group, such as an ⁇ -haloacetic acid, for example, ⁇ - chloroacetic acid, ⁇ -bromoacetic acid, or ⁇ -iodoacetic acid.
  • a linker L ⁇ moiety contains two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety
  • the linker may be conjugated to a solid support Thereafter, two peptide monomers may be synthesized directly onto the two reactive nitrogen groups of the linker L ⁇ moiety in a variation of the solid phase synthesis technique
  • a linker may be conjugated to the two peptide monomers of a peptide dimer after peptide synthesis Such conjugation may be achieved by methods well established in the art.
  • the linker contains at least two functional groups suitable for attachment to the target functional groups of the synthesized peptide monomers For example, a linker with two free amine groups may be reacted with the C-terminal carboxyl groups of each of two peptide monomers.
  • linkers containing two carboxyl groups may be reacted with the N-terminal or side chain amine groups, or C-termmal lysine amides, of each of two peptide monomers
  • the spacer may be incorporated into the peptide during peptide synthesis.
  • a spacer contains a free amino group and a second functional group (e g , a carboxyl group or an ammo group) that enables binding to another molecular moiety
  • the spacer may be conjugated to the solid support. Thereafter, the peptide may be synthesized directly onto the spacer's free amino group by standard solid phase techniques.
  • a spacer containing two functional groups is first coupled to the solid support via a first functional group.
  • a linker L ⁇ moiety having two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety is conjugated to the spacer via the spacer's second functional group and the linker's third functional group
  • two peptide monomers may be synthesized directly onto the two reactive nitrogen groups of the linker L ⁇ moiety in a variation of the solid phase synthesis technique.
  • a solid support coupled spacer with a free amine group may be reacted with a lysine linker via the linker's free carboxyl group.
  • the linker contains at least one functional group suitable for attachment to the target functional group of the synthesized peptide.
  • a spacer with a free amine group may be reacted with a peptide's C-terminal carboxyl group.
  • a linker with a free carboxyl group may be reacted with the free amine group of a peptide's N-terminus or of a lysine residue.
  • a spacer containing a free sulfhydryl group may be conjugated to a cysteine residue of a peptide by oxidation to form a disulfide bond
  • water-soluble polymers such as polyethylene glycol (PEG)
  • PEG polyethylene glycol
  • Attachment of such polymers is thought to enhance biological activity, prolong blood circulation time, reduce lmmunogenicity, increase aqueous solubility, and enhance resistance to protease digestion.
  • PEG polyethylene glycol
  • covalent attachment of PEG to therapeutic polypeptides such as mterleukins [Knauf, et al.
  • the peptide compounds of the invention may further comprise one or more water soluble polymer moieties Preferably, these polymers are covalently attached to the peptide compounds.
  • the water soluble polymer may be, for example, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrohdone, poly-l,3-dioxolane, poly-l,3,6-t ⁇ oxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, polypropylene oxide/ethylene oxide copolymers, and polyoxyethylated polyols.
  • PEG polyethylene glycol
  • copolymers of ethylene glycol/propylene glycol carboxymethylcellulose
  • dextran polyvinyl alcohol
  • polyvinyl pyrrohdone poly-l,3-dioxolane
  • Peptides, peptide dimers and other peptide -based molecules of the invention can be attached to water-soluble polymers (e.g., PEG) using any of a variety of chemistries to link the water-soluble polymer(s) to the receptor-binding portion of the molecule (e.g , peptide + spacer).
  • a typical embodiment employs a single attachment junction for covalent attachment of the water soluble polymer(s) to the receptor-binding portion, however in alternative embodiments multiple attachment junctions may be used, including further variations wherein different species of water-soluble polymer are attached to the receptor-bindmg portion at distinct attachment junctions, which may include covalent attachment junction(s) to the spacer and/or to one or both peptide chains.
  • the dimer or higher order multimer will comprise distinct species of peptide chain (i e., a heterodimer or other heteromul timer).
  • a dimer may comprise a first peptide chain having a PEG attachment junction and the second peptide chain may either lack a PEG attachment junction or utilize a different linkage chemistry than the first peptide chain and in some variations the spacer may contain or lack a PEG attachment junction and said spacer, if PEGylated, may utilize a linkage chemistry different than that of the first and/or second peptide chains.
  • An alternative embodiment employs a PEG attached to the spacer portion of the receptor- binding portion and a different water-soluble polymer (e.g., a carbohydrate) conjugated to a side chain of one of the amino acids of the peptide portion of the molecule.
  • a PEG attached to the spacer portion of the receptor- binding portion and a different water-soluble polymer (e.g., a carbohydrate) conjugated to a side chain of one of the amino acids of the peptide portion of the molecule.
  • PEG polyethylene glycol
  • Suitable reactive PEG species include, but are not limited to, those which are available for sale in the Drug Delivery Systems catalog (2003) of NOF Corporation (Yebisu Garden Place Tower, 20-3 Ebisu 4-chome, Shibuya-ku, Tokyo 150- 6019) and the Molecular Engineering catalog (2003) of Nektar Therapeutics (490 Discovery Drive, Huntsville, Alabama 35806)
  • the following PEG reagents are often preferred in various embodiments: mPEG2-NHS, mPEG2-ALD, multi-Arm PEG, mPEG(MAL)2, mPEG2(MAL), mPEG-NH2, mPEG-SPA, mPEG-SBA, mPEG-thioesters, mPEG-Double Esters, mPEG-BTC, mPEG-ButyrALD, mPEG-ACET, heterofunctional PEGs (NH2-PEG-COOH, Boc- PEG-NHS, Fmoc-PEG-NHS, NHS-
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • a preferred PEG for use in the present invention comprises linear, unbranched PEG having a molecular weight of from about 20 kilodaltons (kD or kDa) to about 4OkD (the term "about” indicating that in preparations of PEG, some molecules will weigh more, some less, than the stated molecular weight).
  • the PEG has a molecular weight of from about 3OkD to about 4OkD.
  • Other sizes may be used, depending on the desired therapeutic profile (e g , duration of sustained release desired; effects, if any, on biological activity; ease in handling; degree or lack of antigenicity; and other known effects of PEG on a therapeutic peptide).
  • the number of polymer molecules attached may vary; for example, one, two, three, or more water soluble polymers may be attached to an EPO-R agonist peptide of the invention.
  • the multiple attached polymers may be the same or different chemical moieties (e g , PEGs of different molecular weight)
  • the degree of polymer attachment (the number of polymer moieties attached to a peptide and/or the total number of peptides to which a polymer is attached) may be influenced by the proportion of polymer molecules versus peptide molecules in an attachment reaction, as well as by the total concentration of each in the reaction mixture.
  • the optimum polymer versus peptide ratio in terms of reaction efficiency to provide for no excess unreacted peptides and/or polymer moieties
  • the desired degree of polymer attachment e g , mono, di-, tri-, etc.
  • the molecular weight of the polymer selected whether the polymer is branched or unbranched, and the reaction conditions for a particular attachment method.
  • the covalently attached water soluble polymer is PEG.
  • PEG PEGylation
  • peptide compounds to which any of a number of water soluble polymers known in the art have been attached by any of a number of attachment methods known in the art are encompassed by the present invention.
  • PEG may serve as a linker that dime ⁇ zes two peptide monomers.
  • PEG is attached to at least one terminus (N-termmus or C-terminus) of a peptide monomer or dimer.
  • PEG is attached to a spacer moiety of a peptide monomer or dimer.
  • PEG is attached to the linker moiety of a peptide dimer.
  • PEG is attached to a spacer moiety, where said spacer moiety is attached to the linker L ⁇ moiety that connects the monomers of a peptide dimer.
  • PEG is attached to a spacer moiety, where said spacer moiety is attached to a peptide dimer via the carbonyl carbon of a lysine linker, or the amide nitrogen of a lysine amide linker.
  • PEG attachment methods available to those skilled in the art [see, e g ,
  • PEG may be covalently bound to ammo acid residues via a reactive group.
  • Reactive groups are those to which an activated PEG molecule may be bound (e g , a free amino or carboxyl group).
  • N-terminal amino acid residues and lysine (K) residues have a free amino group; and C-terminal ammo acid residues have a free carboxyl group.
  • Sulfhydryl groups e g , as found on cysteine residues
  • PEG molecules may be attached to peptide amino groups using methoxylated PEG ("mPEG”) having different reactive moieties.
  • mPEG methoxylated PEG
  • Such polymers include mPEG-succinimidyl succinate, mPEG-succmimidyl carbonate, mPEG-imidate, mPEG-4-nitrophenyl carbonate, and mPEG-cyanu ⁇ c chloride.
  • PEG molecules may be attached to peptide carboxyl groups using methoxylated PEG with a free amine group (mPEG-NH 2 )
  • the desired PEGylated compound may be purified from the mixture of PEGylated compounds.
  • the N-terminally PEGylated form may be purified from a population of randomly PEGylated peptides ( ⁇ e , separating this moiety from other monoPEGylated moieties).
  • PEG is attached site-specifically to a peptide.
  • Site-specific PEGylation at the N-terminus, side chain, and C-terminus of a potent analog of growth hormone- releasing factor has been performed through solid-phase synthesis [Felix, et al. (1995) Int. J. Peptide Protein Res 46'253].
  • Another site-specific method involves attaching a peptide to extremities of liposomal surface-grafted PEG chains in a site-specific manner through a reactive aldehyde group at the N-terminus generated by sodium pe ⁇ odate oxidation of N-terminal threonine [Zahpsky, et al. (1995) Bioconj Chem.
  • selective N-termmal PEGylation may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for de ⁇ vatization in a particular protein. Under the appropriate reaction conditions, a carbonyl group containing PEG is selective attached to the N-termmus of a peptide.
  • PEGylation takes place predominantly at the N-termmus of the protein, with no significant modification of other reactive groups ⁇ e g , lysine side chain amino groups).
  • the PEG should have a single reactive aldehyde for coupling to the protein (e g , PEG prop ⁇ onaldehyde may be used).
  • Site-specific mutagenesis is a further approach which may be used to prepare peptides for site-specific polymer attachment.
  • the ammo acid sequence of a peptide is designed to incorporate an appropriate reactive group at the desired position within the peptide.
  • WO 90/12874 describes the site-directed PEGylation of proteins modified by the insertion of cysteine residues or the substitution of other residues for cysteine residues.
  • This publication also describes the preparation of mPEG-erythropoietin ("mPEG-EPO”) by reacting a cysteme-specific mPEG derivative with a recombinantly introduced cysteine residue on EPO.
  • mPEG-EPO mPEG-erythropoietin
  • the linker or spacer contains a reactive group and an activated PEG molecule containing the approp ⁇ ate complementary reactive group is used to effect covalent attachment.
  • the linker or spacer reactive group contains a terminal amino group (/ e , positioned at the terminus of the linker or spacer) which is reacted with a suitably activated PEG molecule to make a stable covalent bond such as an amide or a carbamate.
  • Suitable activated PEG species include, but are not limited to, mPEG-para-nitrophenylcarbonate (mPEG-NPC), mPEG-succinimidyl carbonate (mPEG-SC), and mPEG-succinimidyl propionate (mPEG-SPA).
  • the linker or spacer reactive group contains a carboxyl group capable of being activated to form a covalent bond with an amine-containmg PEG molecule under suitable reaction conditions.
  • Suitable PEG molecules include mPEG-NB ⁇ and suitable reaction conditions include carbodnmide-mediated amide formation or the like.
  • the biological activity of the various peptide compounds of this invention can be assayed by any of a variety of methods that are well known in the art. See, for example, in International Patent Application No. PCT/US04/14886, filed May 12, 2004 Non-hmiting examples of certain, preferred assays are also described here.
  • EPO binding protein EBP
  • EBP extracellular domain of the human EPO-R
  • a solid support such as a microtitre dish or a synthetic bead [e g , Sulfolink beads from Pierce Chemical Co. (Rockford, IL)].
  • Immobilized EBP is then incubated with labeled recombinant EPO, or with labeled recombinant EPO and a test peptide Serial dilutions of test peptide are employed for such experiments.
  • Assay points with no added test peptide define total EPO binding to EBP.
  • the IC50 value is defined as the concentration of test peptide which reduces the binding of EPO to EBP by 50% ( ⁇ e , 50% inhibition of EPO binding).
  • a different in vitro competitive binding assay measures the light signal generated as a function of the proximity of two beads: an EPO-conjugated bead and an EPO-R-conjugated bead. Bead proximity is generated by the binding of EPO to EPO-R. A test peptide that competes with EPO for binding to EPO-R will prevent this binding, causing a decrease in light emission The concentration of test peptide that results in a 50% decrease in light emission is defined as the IC50 value
  • the biological activity and potency of monomeric and dimeric peptide EPO-R agonists of the invention, which bind specifically to the EPO-receptor, may be measured using in vitro cell-based functional assays
  • One assay is based upon a murine pre-B-cell line expressing human EPO-R and further transfected with a fos promoter-driven luciferase reporter gene construct.
  • Luciferase causes the emission of light upon addition of its substrate lucife ⁇ n
  • the level of EPO-R activation in such cells may be quantitated via measurement of luciferase activity
  • the activity of a test peptide is measured by adding serial dilutions of the test peptide to the cells, which are then incubated for 4 hours. After incubation, lucife ⁇ n substrate is added to the cells, and light emission is measured. The concentration of test peptide that results in a half-maximal emission of light is recorded as the EC50
  • Another assay may be performed using FDC-P1/ER cells [Dexter, et al. (1980) J. Exp Med 152 1036-1047], a well characterized nontransformed murine bone marrow derived cell line into which EPO-R has been stably transfected These cells exhibit EPO-dependent proliferation.
  • the cells are grown to half stationary density in the presence of the necessary growth factors (see, e g , as described in U.S. Patent 5,773,569). The cells are then washed in PBS and starved for 16-24 hours in whole media without the growth factors.
  • Cell proliferation is then measured by techniques known m the art, such as an MTT assay which measures H 3 -thymidme incorporation as an indication of cell proliferation [see, Mosmann (1983) J Immunol Methods 65-55-63].
  • Peptides are evaluated on both the EPO-R-expressing cell line and a parental non-expressing cell line. The concentration of test peptide necessary to yield one half of the maximal cell proliferation is recorded as the EC50.
  • Li another assay the cells are grown to stationary phase m EPO-supplemented medium, collected, and then cultured for an additional 18 hr in medium without EPO.
  • the cells are divided into three groups of equal cell density: one group with no added factor (negative control), a group with EPO (positive control), and an experimental group with the test peptide.
  • the cultured cells are then collected at various time points, fixed, and stained with a DNA-binding fluorescent dye (e g , propidium iodide or Hoechst dye, both available from Sigma). Fluorescence is then measured, for example, using a FACS Scan Flow cytometer.
  • a DNA-binding fluorescent dye e g , propidium iodide or Hoechst dye, both available from Sigma. Fluorescence is then measured, for example, using a FACS Scan Flow cytometer.
  • the percentage of cells m each phase of the cell cycle may then be determined, for example, using the SOBR model of CeIIFIT software (Becton Dickinson). Cells treated with EPO or an active peptide will show a greater proportion of cells in S phase (as determined by increased fluorescence as an indicator of increased DNA content) relative to the negative control group.
  • FDCP-I is a growth factor dependent murine multi-potential primitive hematopoietic progenitor cell line that can proliferate, but not differentiate, when supplemented with WEHI-3 -conditioned media (a medium that contains IL-3, ATCC number TEB-68).
  • the FDCP-I cell line is transfected with the human or murine EPO-R to produce FDCP- 1-hEPO-R or FDCP- 1 -mEPO-R cell lines, respectively, that can proliferate, but not differentiate, in the presence of EPO.
  • TF-I an EPO- dependent cell line, may also be used to measure the effects of peptide EPO-R agonists on cellular proliferation.
  • mice are injected daily for two days with phenylhydrazine (60 mg/kg). On the third day, spleen cells are removed and their ability to proliferate over a 24 hour period ascertained using an MTT assay.
  • EPO-R The binding of EPO to EPO-R in an erythropoietin-responsive cell line induces tyrosine phosphorylation of both the receptor and numerous intracellular proteins, including She, vav and JAK2 kinase. Therefore, another in vitro assay measures the ability of peptides of the invention to induce tyrosine phosphorylation of EPO-R and downstream intracellular signal transducer proteins. Active peptides, as identified by binding and proliferation assays described above, elicit a phosphorylation pattern nearly identical to that of EPO in erythropoietin-responsive cells. For this assay, FDC-P1/ER cells [Dexter, et al.
  • the blot may be probed with an anti-phosphotyrosme antibody (e g , mouse anti -phosphotyrosine IgG from Upstate Biotechnology, Inc.), washed, and then probed with a secondary antibody [e g , peroxidase labeled goat anti-mouse IgG from Kirkegaard & Perry Laboratories, Inc. (Washington, DC)].
  • an anti-phosphotyrosme antibody e g , mouse anti -phosphotyrosine IgG from Upstate Biotechnology, Inc.
  • a secondary antibody e g , peroxidase labeled goat anti-mouse IgG from Kirkegaard & Perry Laboratories, Inc. (Washington, DC)
  • phosphotyrosine-containing proteins may be visualized by standard techniques including colo ⁇ met ⁇ c, chemiluminescent, or fluorescent assays.
  • a chemiluminescent assay may be performed using the ECL Western Blotting System from Amersham
  • Murine bone marrow may be obtained from the femurs of mice, while a sample of human peripheral blood may obtained from a healthy donor.
  • mononuclear cells are first isolated from the blood, for example, by centrifugation through a Ficoll-Hypaque gradient [Stem Cell Technologies, Inc. (Vancouver, Canada)]. For this assay a nucleated cell count is performed to establish the number and concentration of nucleated cells in the original sample.
  • a defined number of cells is plated on methyl cellulose as per manufacturer's instructions [Stem Cell Technologies, Inc. (Vancouver, Canada)].
  • An experimental group is treated with a test peptide, a positive control group is treated with EPO, and a negative control group receives no treatment.
  • the number of growing colonies for each group is then scored after defined periods of incubation, generally 10 days and 18 days. An active peptide will promote colony formation.
  • Biol Chem 267.17055-17060 tyrosine phosphorylation of a cytosohc protein, pp 100, in FDC-ER cells
  • Worthington, et al. (1987) Exp. Hematol. 15:85-92 colon ⁇ metric assay for hemoglobin
  • Kaiho and Miuno (1985) Anal. Biochem. 149: 117-120 detection of hemoglobin with 2,7-diaminofluorene
  • Patel et al. (1992) J. Biol. Chem. 267:21300- 21302 (expression of c-myb); Witthuhn, et al.
  • microphysiometer An instrument designed by Molecular Devices Corp., known as a microphysiometer, has been reported to be successfully used for measurement of the effect of agonists and antagonists on various receptors.
  • the basis for this apparatus is the measurement of the alterations in the acidification rate of the extracellular media in response to receptor activation.
  • mice are subjected to an alternating conditioning cycle for several days. In this cycle, the mice alternate between periods of hypobaric conditions and ambient pressure conditions. Thereafter, the mice are maintained at ambient pressure for 2-3 days prior to administration of test samples
  • Test peptide samples or EPO standard m the case positive control mice, are injected subcutaneously into the conditioned mice. Radiolabeled iron (e g , Fe 59 ) is administered 2 days later, and blood samples taken two days after administration of radiolabeled iron.
  • reticulocyte assay Another in vivo functional assay that may be used to assess the potency of a test peptide is the reticulocyte assay.
  • normal untreated mice are subcutaneously injected on three consecutive days with either EPO or test peptide. On the third day, the mice are also intraperitoneally injected with iron dextran. At day five, blood samples are collected from the mice. The percent (%) of reticulocytes in the blood is determined by thiazole orange staining and flow cytometer analysis (retic-count program). In addition, hematocrits are manually determined. The percent of corrected reticulocytes is determined using the following formula:
  • % RETICcORRECTED % RETICoBSERVED X (Hemat ⁇ C ⁇ t, ND ,viDUAL /
  • the peptide compounds of the invention are useful in vitro as tools for understanding the biological role of EPO, including the evaluation of the many factors thought to influence, and be influenced by, the production of EPO and the binding of EPO to the EPO-R (e g , the mechanism of EPO / EPO-R signal transduction/receptor activation).
  • the present peptides are also useful in the development of other compounds that bind to the EPO-R, because the present compounds provide important structure-activity-relationship information that facilitate that development.
  • the peptides of the present invention can be used as reagents for detecting EPO-R on living cells; fixed cells; m biological fluids; in tissue homogenates; in purified, natural biological materials; etc. For example, by labeling such peptides, one can identify cells having EPO-R on their surfaces.
  • the peptides of the present invention can be used in in situ staining, FACS (fluorescence-activated cell sorting) analysis, Western blotting, ELISA (enzyme-linked immunosorbent assay), etc.
  • the peptides of the present invention can be used m receptor purification, or in purifying cells expressing EPO-R on the cell surface (or inside permeabihzed cells)
  • the peptides of the invention can also be utilized as commercial reagents for various medical research and diagnostic purposes. Such uses can include but are not limited to: (1) use as a calibration standard for quantitating the activities of candidate EPO-R agonists in a variety of functional assays; (2) use as blocking reagents in random peptide screening, i.e , in looking for new families of EPO-R peptide hgands, the peptides can be used to block recovery of EPO peptides of the present invention; (3) use in co-crystalhzation with EPO-R, i e , crystals of the peptides of the present invention bound to the EPO-R may be formed, enabling determination of receptor/peptide structure by X-ray crystallography; (4) use to measure the capacity of erythrocyte precursor cells induce globm synthesis and heme complex synthesis, and to increase the number of ferritin receptors, by initiating differentiation; (5) use to maintain the proliferation and growth of EPO-
  • peptide compounds of the invention may be administered to warm blooded animals, including humans, to simulate the binding of EPO to the EPO-R in vivo.
  • the present invention encompasses methods for therapeutic treatment of disorders associated with a deficiency of EPO, which methods comprise administering a peptide of the invention in amounts sufficient to stimulate the EPO-R and thus, alleviate the symptoms associated with a deficiency of EPO in vivo.
  • the peptides of this invention will find use in the treatment of renal insufficiency and/or end-stage renal failure/dialysis; anemia associated with AIDS; anemia associated with chronic inflammatory diseases (for example, rheumatoid arthritis and chronic bowel inflammation) and autoimmune disease; and for boosting the red blood count of a patient prior to surgery
  • Other disease states, disorders, and states of hematologic irregularity that may be treated by administration of the peptides of this invention include, beta-thalassemia; cystic fibrosis; pregnancy and menstrual disorders, early anemia of prematurity; spinal cord injury, space flight; acute blood loss, aging, and various neoplastic disease states accompanied by abnormal erythropoiesis.
  • the peptide compounds of the invention may be used for the treatment of disorders which are not characterized by low or deficient red blood cells, for example as a pretreatment prior to transfusions.
  • administration of the compounds of this invention can result in a decrease in bleeding time and thus, will find use m the administration to patients prior to surgery or for indications wherein bleeding is expected to occur.
  • the compounds of this invention will find use in the activation of megakaryoctes.
  • EPO has been shown to have a mitogenic and chemotactic effect on vascular endothelial cells as well as an effect on central cholinergic neurons [see, e g , Amagnostou, et al. (1990) Proc. Natl. Acad. Sci USA 87:5978-5982 and Konishi, et al. (1993) Brain Res. 609:29-35], the compounds of this invention will also find use for the treatment of a variety of vascular disorders, such as. promoting wound healing; promoting growth of collateral coronary blood vessels (such as those that may occur after myocardial infarction); trauma treatment; and post-vascular graft treatment.
  • vascular disorders such as. promoting wound healing; promoting growth of collateral coronary blood vessels (such as those that may occur after myocardial infarction); trauma treatment; and post-vascular graft treatment.
  • the compounds of this invention will also find use for the treatment of a variety of neurological disorders, generally characterized by low absolute levels of acetyl choline or low relative levels of acetyl choline as compared to other neuroactive substances e g., neurotransmitters.
  • compositions of the above EPO-R agonist peptide compounds are provided. Conditions alleviated or modulated by the administration of such compositions include those indicated above.
  • Such pharmaceutical compositions may be for administration by oral, parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), transdermal (either passively or using iontophoresis or electroporation), transmucosal (nasal, vaginal, rectal, or sublingual) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate for each route of administration.
  • compositions comprising effective amounts of an EPO-R agonist peptide, or derivative products, of the invention together with pharmaceutically acceptable diluents, preservatives, solubihzers, emulsifiers, adjuvants and/or carriers.
  • compositions include diluents of various buffer content ⁇ eg , T ⁇ s-HCl, acetate, phosphate), pH and ionic strength, additives such as detergents and solubilizing agents ⁇ e.g , Tween 20, Tween 80, Polysorbate 80), anti-oxidants (e g , ascorbic acid, sodium metabisulfite), preservatives (e g , Thimersol, benzyl alcohol) and bulking substances (e g , lactose, manmtol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc or into liposomes Hylauronic acid may also be used Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present proteins and derivatives.
  • additives such as detergents and solubilizing agents ⁇ e.g , Tween 20, Tween 80, Polysorbate 80
  • compositions may be prepared in liquid form, or may be in dried powder (e g , lyophihzed) form.
  • Solid dosage forms include tablets, capsules, pills, troches or lozenges, cachets, pellets, powders, or granules
  • liposomal or protemoid encapsulation may be used to formulate the present compositions (as, for example, protemoid microspheres reported in U S Patent No 4,925,673) Liposomal encapsulation may be used and the liposomes may be de ⁇ vatized with various polymers (e g , U S Patent No.
  • the formulation will include the EPO-R agonist peptides (or chemically modified forms thereof) and inert ingredients which allow for protection against the stomach environment, and release of the biologically active material in the intestine.
  • liquid dosage forms for oral administration including pharmaceutically acceptable emulsions, solutions, suspensions, and syrups, which may contain other components including inert diluents; adjuvants such as wetting agents, emulsifying and suspending agents; and sweetening, flavoring, and perfuming agents.
  • the peptides may be chemically modified so that oral delivery of the derivative is efficacious.
  • the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of proteolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the component or components and increase in circulation time in the body.
  • PEGylation is a preferred chemical modification for pharmaceutical usage
  • Other moieties that may be used include: propylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrohdone, polyprohne, poly-l ,3-dioxolane and poly-l,3,6-tioxocane [see, e g , Abuchowski and Davis (1981) "Soluble Polymer-Enzyme Adducts," in Enzymes as Drugs Hocenberg and Roberts, eds (Wiley-Interscience: New York, NY) pp. 367-383; and Newmark, et al (1982) J Appl. Biochem. 4- 185-189]
  • the location of release may be the stomach, the small intestine (the duodenum, the jejunem, or the ileum), or the large intestine
  • the release will avoid the deleterious effects of the stomach environment, either by protection of the peptide (or derivative) or by release of the peptide (or derivative) beyond the stomach environment, such as in the intestine.
  • a coating impermeable to at least pH 5.0 is essential.
  • Examples of the more common inert ingredients that are used as enteric coatings are cellulose acetate t ⁇ melhtate (CAT), hydroxypropylmethylcellulose phthalate (HPMCP), HPMCP 50, HPMCP 55, polyvinyl acetate phthalate (PVAP), Eudragit L30D, Aquate ⁇ c, cellulose acetate phthalate (CAP), Eudragit L, Eudragit S, and Shellac. These coatings may be used as mixed films. A coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach.
  • Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic ( ⁇ e powder), for liquid forms a soft gelatin shell may be used.
  • the shell mate ⁇ al of cachets could be thick starch or other edible paper.
  • moist massing techniques can be used.
  • the peptide (or derivative) can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about lmm.
  • the formulation of the material for capsule administration could also be as a powder, lightly compressed plugs, or even as tablets. These therapeutics could be prepared by compression.
  • Colorants and/or flavoring agents may also be included.
  • the peptide (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.
  • diluents could include carbohydrates, especially mannitol, ⁇ -lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch
  • Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride.
  • Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell.
  • Disintegrants may be included in the formulation of the therapeutic into a solid dosage form.
  • Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberhte, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used.
  • the disintegrants may also be insoluble cationic exchange resins.
  • Powdered gums may be used as disintegrants and as binders, and can include powdered gums such as agar, Karaya or tragacanth.
  • Binders may be used to hold the peptide (or derivative) agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC) Polyvinyl pyrrolidone (PVP) and hydroxypropyl methyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the peptide (or derivative).
  • MC methyl cellulose
  • EC ethyl cellulose
  • CMC carboxymethyl cellulose
  • PVP polyvinyl pyrrolidone
  • HPMC hydroxypropyl methyl cellulose
  • An antifrictional agent may be included in the formulation of the peptide (or derivative) to prevent sticking during the formulation process.
  • Lubricants may be used as a layer between the peptide (or derivative) and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
  • Ghdants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added.
  • the ghdants may include starch, talc, pyrogenic silica and hydrated silicoalummate
  • surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
  • Cationic detergents might be used and could include benzalkomum chloride or benzethomium chloride.
  • nonionic detergents that could be included in the formulation as surfactants are lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 20, 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose
  • surfactants could be present in the formulation of the protein or derivative either alone or as a mixture in different ratios
  • Additives which potentially enhance uptake of the peptide (or derivative) are for instance the fatty acids oleic acid, linoleic acid and linolenic acid
  • Controlled release oral formulations may be desirable
  • the peptide (or derivative) could be incorporated into an inert matrix which permits release by either diffusion or leaching mechanisms, e g , gums
  • Slowly degenerating matrices may also be incorporated into the formulation.
  • Some enteric coatings also have a delayed release effect
  • Another form of a controlled release is by a method based on the Oros therapeutic system (Alza Corp.), i e the drug is enclosed in a semipermeable membrane which allows water to enter and push drug out through a single small opening due to osmotic effects.
  • the peptide (or derivative) could also be given in a film coated tablet and the materials used in this instance are divided into 2 groups
  • the first are the nonente ⁇ c materials and include methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl -methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols.
  • the second group consists of the enteric materials that are commonly esters of phthahc acid.
  • Film coating may be carried out in a pan coater or in a fluidized bed or by compression coating.
  • Preparations according to this invention for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions.
  • non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate.
  • Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use.
  • compositions for rectal or vaginal administration are preferably suppositories which may contain, in addition to the active substance, excipients such as cocoa butter or a suppository wax.
  • compositions for nasal or sublingual administration are also prepared with standard excipients well known in the art.
  • the peptide (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream [see, e g , Adjei, et al. (1990) Pharmaceutical Research 7:565-569; Adjei, et al. (1990) Int. J Pharmaceutics 63: 135-144 (leuprohde acetate); Braquet, et al. (1989) J.
  • each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy.
  • propellant material in addition to the usual diluents, adjuvants and/or carriers useful in therapy.
  • liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated
  • Chemically modified peptides may also be prepared in different formulations depending on the type of chemical modification or the type of device employed
  • Formulations suitable for use with a nebulizer will typically comprise peptide (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active protein per mL of solution
  • the formulation may also include a buffer and a simple sugar (e g , for protein stabilization and regulation of osmotic pressure).
  • the nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the peptide (or derivative) caused by atomization of the solution in forming the aerosol
  • Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the peptide (or derivative) suspended in a propellant with the aid of a surfactant.
  • the propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including t ⁇ chlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and
  • Suitable surfactants include sorbitan trioleate and soya lecithin Oleic acid may also be useful as a surfactant.
  • Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing peptide (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e g , 50 to 90% by weight of the formulation.
  • the peptide (or derivative) should most advantageously be prepared in particulate form with an average particle size of less than 10 mm (or microns), most preferably 0.5 to 5 mm, for most effective delivery to the distal lung.
  • Nasal delivery of the EPO-R agonist peptides is also contemplated.
  • Nasal delivery allows the passage of the peptide to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung
  • Formulations for nasal delivery include those with dextran or cyclodextran.
  • Other penetration-enhancers used to facilitate nasal delivery are also contemplated for use with the peptides of the present invention (such as described in International Patent Publication No. WO 2004056314, filed December 17, 2003, incorporated herein by reference in its entirety).
  • peptide compounds For all of the peptide compounds, as further studies are conducted, information will emerge regarding appropriate dosage levels for treatment of various conditions in various patients, and the ordinary skilled worker, considering the therapeutic context, age, and general health of the recipient, will be able to ascertain proper dosmg
  • the selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. Generally dosage levels of 0.001 to 10 mg/kg of body weight daily are administered to mammals. Generally, for intravenous injection or infusion dosage may be lower.
  • the dosing schedule may vary, depending on the circulation half-life, and the formulation used.
  • the peptides of the present invention may be administered in conjunction with one or more additional active ingredients or pharmaceutical compositions.
  • Example 1 Synthesis of EPO-R agonist peptides This example describes preferred, non-limiting embodiments of methods by which peptides covered by the present invention can be synthesiaed. However, other methods, which have been previously described for the synthesis of EPO peptide moieties (see, for example, in PCT/US04/14886, filed May 12, 2004) can also be used to prepare compounds of this invention.
  • Solid phase techniques are provided for synthesizing both peptide monomers and dimers of the invention.
  • Exemplary techniques for attaching linker and PEG moieties to a peptide compound of this invention are also described, as well as methods for oxidizing the peptide compounds, e.g., forming intramolecular disulfide bonds.
  • this example also provides a technique for purifying peptide compounds that are synthesized according to these methods.
  • PAL resm results in a carboxyl terminal amide functional group upon cleavage of the peptide from the resin
  • Primary amine protection on amino acids is achieved with Fmoc, and side chain protection groups is t-butyl for serine, threonine, and tyrosine hydroxyls; t ⁇ tyl for glutamine and asparagme amides; Trt or Acm for cysteine; and PMC (2,2,5,7,8-pentamethylchroman sulfonate) for the argimne guanidmo group.
  • Each coupling is performed for either 1 hr or 2 hr with BOP (benzotriazolyl N- oxt ⁇ sdimethylammophosphonium hexafluorophosphate) and HOBt (1 -hydroxybenztriazole).
  • BOP benzotriazolyl N- oxt ⁇ sdimethylammophosphonium hexafluorophosphate
  • HOBt 1-hydroxybenztriazole
  • the fully assembled peptide is cleaved with a mixture of 90% trifluoroacetic acid, 5% ethanedithiol, and 5% water, initially at 4°C and gradually increased to room temperature over 1.5 hr.
  • the deprotected product is filtered from the resm and precipitated with diethyl ether. After thorough drying the product is purified by Cl 8 reverse phase high performance liquid chromatography with a gradient of acetonit ⁇ le/water in 0 1% trifluoroacetic acid.
  • peptide dimers of the invention are synthesized directly onto a lysine linker in a variation of the solid phase technique.
  • Fmoc-Lys(Fmoc)-OH is coupled to a PAL resin (Milhgen/Biosearch), thereby providing an initial lysine residue to serve as the linker between the two chains to be synthesized
  • the Fmoc protecting groups are removed with mild base (20% pipe ⁇ dme in DMF), and the peptide chains are synthesized using the resulting free ammo groups as starting points.
  • Peptide chain synthesis is performed using the solid phase synthesis technique described above. Trt is used to protect all cysteine residues.
  • oxidation of the cysteine residues is performed by incubating the deprotected dimer in 100% DMSO for 2-3 days at 5°C to 25°C. This oxidation reaction can yield predominantly (>75%) dimers with two intramolecular disulfide bonds.
  • Fmoc-Lys(AUoc)-OH is coupled to a PAL resin (Milhgen/Biosearch), thereby providing an initial lysine residue to serve as the linker between the two chains to be synthesized.
  • the Fmoc protecting group is removed with mild base (20% pipe ⁇ dine in DMF)
  • the first peptide chain is then syntheszed using the resulting free amino group as a starting point Peptide synthesis is performed using the solid phase technique described above
  • the two cysteine residues of the first chain are protected with Trt
  • the Alloc group is removed from the support-bound lysine linker with Pd[P(C 6 H 5 ) 3 ] 4 , 4-methyl morphohne, and chloroform
  • the second peptide chain is then synthesized on this second free amino group
  • the two cysteine residues of the second chain are protected with Acm
  • An intramolecular disulfide bond is formed in the first peptide chain by removing the Trt protecting groups using t ⁇ fluoroacetic acid, followed by oxidation by stirring in 20% DMSO overnight
  • An intramolecular disulfide bond is then formed m the second peptide chain by simultaneously removing the Acm protecting groups and oxid
  • the spacer is an ammo acid (e g , glycine or lysine)
  • the spacer is incorporated into the peptide during solid phase peptide synthesis
  • the spacer amino acid is coupled to the PAL resin, and its free ammo group can serve as the basis for the attachment of another spacer ammo acid, or of the lysine linker Following the attachment of the lysine linker, dimeric peptides are synthesized as described above
  • the peptide dimer is dissolved in 20% DMSO/water (1 mg dry weight peptide/mL) and is allowed to stand at room temperature for 36h
  • the peptide is purified by loading the reaction mixture onto a Cl 8 HPLC column (Waters Delta-Pak Cl 8, 15 micron particle size, 300 angstrom pore size, 40 mm x 200 mm length), followed by a linear ACN/water/0 01% TFA gradiant from 5 to 95% ACN over 40 minutes Lyophohzation of the fractions containing the desired peptide affords the product as a fluffy white solid
  • PEGylation of a terminal -NH 2 group The peptide dimer is mixed with 1 5 eq (mole basis) of activated PEG species (mPEG-NPC from NOF Corp Japan) in dry DMF to afford a clear solution
  • Either the peptide prior to conjugation or the peptide-PEG conjugate is dissolved in 25% ACN/water (10 mg/mL) and the pH adjusted to ⁇ 3 with TFA, then loaded on the column. After washing with 2-3 column volumes of 25% ACN/water and collecting 5 mL fractions, the peptide is released from the column by elution with 0 1 M NH 4 OAc in 25% ACN/water, again collecting 5 mL fractions. Analysis via HPLC can reveal which fractions contain the desired peptide.
  • Evaporative Light- Scattering Detector can indicate that when the peptide is retained on the column and is eluted with the NH 4 OAc solution (generally between fractions 4 and 10), no non-conjugated PEG is observed as a contaminant.
  • the peptide elutes in the initial wash buffer (generally the first 2 fractions), no separation of desired PEG-conjugate and excess PEG may be observed.
  • This example describes certain in vitro assays that are useful for evaluating the activity and potency of peptides covered by this invention, e.g., as EPO-R agonists.
  • the results obtained from assays such as the ones described here demonstrate whether a peptide compound binds to EPO-R and activates EPO-R signalling.
  • the assays can also be used to compare the binding efficiency and biological activity of a compound, for example, to other, known EPO mimetic compounds
  • EPO-R agonist peptide monomers and dimers tested in these assays are typically prepared according to methods such as those described in Example 1.
  • the potency of these peptide monomers and dimers is then evaluated using a series of in vitro activity assays, including: a reporter assay, a proliferation assay, a competitive binding assay, and a C/BFU-e assay. These four assays are described in further detail below.
  • This assay is based upon a on a murine pre-B-cell line derived reporter cell, Baf3/EpoR/GCSFR fos/lux.
  • This reporter cell line expresses a chimeric receptor comprising the extracellular portion of the human EPO receptor to the lntra-cellular portion of the human GCSF receptor.
  • This cell line is further transfected with a fos promoter-driven luciferase reporter gene construct. Activation of this chimeric receptor through addition of erythropoietic agent results in the expression of the luciferase reporter gene, and therefore the production of light upon addition of the luciferase substrate lucife ⁇ n.
  • the level of EPO-R activation in such cells may be quantitated via measurement of luciferase activity.
  • the Baf3/EpoR/GCSFR fos/lux cells are cultured in DMEM/F12 medium (Gibco) supplemented with 10% fetal bovine serum (FBS; Hyclone), 10% WEHI-3 supernatant (the supernatant from a culture of WEHI-3 cells, ATCC # TIB-68), and penicillin/streptomycin. Appioximately 18 h before the assay, cells are starved by transferring them to DMEM/F12 medium supplemented with 10% FBS and 0.1% WEHI-3 supernatant.
  • FBS fetal bovine serum
  • WEHI-3 supernatant the supernatant from a culture of WEHI-3 cells, ATCC # TIB-68
  • penicillin/streptomycin Appioximately 18 h before the assay, cells are starved by transferring them to DMEM/F12 medium supplemented with 10% FBS and 0.1% WEHI-3 supernatant.
  • test peptide Serial dilutions of the test peptide are concurrently tested in this assay Assay plates are incubated for 4h at 37 0 C in a 5% CO 2 atmosphere, after which lucife ⁇ n (Steady-Glo; Promega, Madison, WI) is added to each well Following a 5-minute incubation, light emission is measured on a Packard Topcount Luminometer (Packard Instrument Co., Downers Grove, 111.)- Light counts are plotted relative to test peptide concentration and analysed using Graph Pad software. The concentration of test peptide that results in a half-maximal emission of light is recorded as the EC50.
  • lucife ⁇ n Steady-Glo; Promega, Madison, WI
  • This assay is based upon a murine pre-B-cell line, BaO, transfected to express human EPO-R. Proliferation of the resulting cell line, BaF3/Gal4/Elk/EPOR, is dependent on EPO-R activation. The degree of cell proliferation is quantitated using MTT, where the signal in the MTT assay is proportional to the number of viable cells.
  • the BaF3/Gal4/Elk/EPOR cells are cultured in spinner flasks m DMEM/F12 medium (Gibco) supplemented with 10% FBS (Hyclone) and 2% WEHI-3 supernatant (ATCC # TBB-68).
  • Cultured cells are starved overnight, in a spmner flask at a cell density of IxIO 6 cells/ml, in DMEM/F12 medium supplemented with 10% FBS and 0.1% WEHI-3 supernatant.
  • the starved cells are then washed twice with Dulbecco's PBS (Gibco), and resuspended to a density of 1x10 6 cells/ml in DMEM/F12 supplemented with 10% FBS (no WEHI-3 supernatant). 50 ⁇ L ahquots (-50,000 cells) of the cell suspension are then plated, in triplicate, in 96 well assay plates.
  • test EPO mimetic peptides 50 ⁇ L ahquots of dilution series of test EPO mimetic peptides, or 50 ⁇ L EPO (R & D Systems Inc., Minneapolis, MN) or AranespTM (darbepoeitm alpha, an ERO-R agonist comme ⁇ cally available from Amgen) in DMEM/F12 media supplemented with 10% FBS (no WEHI-3 supernatant I) are added to the 96 well assay plates (final well volume of lOO ⁇ L). For example, 12 different dilutions may be tested where the final concentration of test peptide (or control EPO peptide) ranges from 81OpM to 0.0045pM. The plated cells are then incubated for 48h at 37 0 C.
  • the method is as follows: Add 4 ⁇ L of serial dilutions of the test EPO-R agonist peptide, or positive or negative controls, to wells of a 384 well plate. Thereafter, add 2 ⁇ L / well of receptor/bead cocktail.
  • Receptor bead cocktail consists of 15 ⁇ L of 5mg/ml streptavidin donor beads (Packard), 15 ⁇ L of 5mg/ml monoclonal antibody abl79 (this antibody recognizes the portion of the human placental alkaline phosphatase protein contained in the recombinant EPO-R), protein A-coated acceptor beads (protein A will bind to the abl79 antibody; Packard), 112.5 ⁇ L of a 1 :6.6 dilution of recombinant EPO-R (produced in Chinese Hamster Ovary cells as a fusion protein to a portion of the human placental alkaline phosphatase protein which contains the abl79 target epitope) and 607.5 ⁇ L of Alphaquest buffer (4OmM HEPES, pH 7.4; ImM MgCl 2 ; 0.1% BSA, 0.05% Tween 20). Tap to mix. Add 2 ⁇ L/well of a biotinylated EPO-R-bmding peptide trace
  • the concentration of test peptide that results in a 50% decrease in light emission, relative to that observed without test peptide, is recorded as the IC50.
  • EPO-R signaling stimulates the differentiation of bone marrow stem cells into proliferating red blood cell presursors.
  • This assay measures the ability of test peptides to stimulate the proliferation and differentiation of red blood cell precursors from primary human bone marrow plu ⁇ potent stem cells.
  • test peptide serial dilutions of test peptide are made in EVIDM medium (Gibco) supplemented with 10% FBS (Hyclone). These serial dilutions, or positive control EPO peptide, are then added to methylcellulose to give a final volume of 1.5mL. The methylcellulose and peptide mixture is then vortexed thoroughly. Ahquots (100,000 cells/mL) of human, bone marrow de ⁇ ved CD34+ cells (Poietics/Cambrex) are thawed. The thawed cells are gently added to 0 1 mL of lmg/ml DNAse (Stem Cells) in a 5OmL tube.
  • EVIDM medium Gibco
  • FBS Hyclone
  • IMDM medium 40-50 mL IMDM medium is added gently to cells: the medium is added drop by drop along the side of the 5OmL tube for the first 1OmL, and then the remaining volume of medium is slowly dispensed along the side of the tube. The cells are then spun at 900rpm for 20 mm, and the media removed carefully by gentle aspiration. The cells are resuspended in ImI of IMDM medium and the cell density per mL is counted on hemacytometer slide (lO ⁇ L aliquot of cell suspension on slide, and cell density is the average count X 10,000 cells/ml). The cells are then diluted in IMDM medium to a cell density of 15,000 cells/mL.
  • a lOO ⁇ L of diluted cells is then added to each 1.5 mL methyl cellulose plus peptide sample (final cell concentration in assay media is 1000 cells/ mL), and the mixture is vortexed. Allow the bubbles in the mixture to disappear, and then aspirate ImL using blunt-end needle.
  • radioligand competition binding assay can also be used to measure IC50 values for peptides of the present invention.
  • This assay measures binding of 125 I-EPO to EPOr.
  • the assay may be performed according to the following exemplary protocol: A. Materials
  • One 50 ⁇ g vial of lyophihzed recombinant EPOR extracellular domain fused to the Fc portion of human IgGl is reconstituted in 1 mL of assay buffer.
  • 100 ⁇ L serial dilutions of this receptor preparation are combined with approximately 20,000 cpm in 200 ⁇ L of iodinated recombinant human Erythropoietin ( 125 I-EPO) in 12 x 75 mm polypropylene test tubes Tubes are capped and mixed gently at 4 0 C overnight on a LabQuake rotating shaker
  • IC 50 of a peptide of the present invention 100 ⁇ L serial dilutions of the peptide are combined with 100 ⁇ L of recombinant erythropoietin receptor (100 pg/tube) in 12 x 75 mm polypropylene test tubes Then 100 ⁇ L of iodmated recombinant human Erythropoietin ( 125 I- EPO) is added to each tube and the tubes were capped and mixed gently at 4 0 C overnight The next day, bound ' 25 I-EPO is quantitated as described above The results are analyzed and the IC 50 value calculated using Graphpad Prism version 4 0, from GraphPad Software, Lie (San Diego, CA) The assay is preferably repeated 2 or more times for each peptide whose IC 50 value is measured by this procedure, for a total of 3 replicate IC 50 determinations
  • This example describes certain in vivo assays that are useful for evaluating the activity and potency of peptides covered by this invention, e g , as EPO-R agonists
  • the results obtained from assays such as the ones described here demonstrate whether a peptide compound binds to EPO-R and activates EPO-R signalling
  • the assays can also be used to compare the binding efficiency and biological activity of a compound, for example, to other, known EPO mimetic compounds
  • EPO-R agonist peptide monomers and dimers tested in these assays are typically prepared according to the methods described in Example 1
  • the in vivo activity of these peptide monomers and dimers is then evaluated using a series assays, including a polycythemic exhypoxic mouse bioassay and a reticulocyte assay.
  • Test peptides are assayed for in vivo activity in the polycythemic exhypoxic mouse bioassay adapted from the method described by Cotes and Bangham (1961), Nature 191 ' 1065-1067.
  • This assay examines the ability of a test peptide to function as an EPO mimetic: i e , to activate EPO-R and induce new red blood cell synthesis Red blood cell synthesis is quantitated based upon incorporation of radiolabeled iron into hemoglobin of the synthesized red blood cells.
  • BDFl mice are allowed to acclimate to ambient conditions for 7-10 days. Body weights are determined for all animals, and low weight animals ( ⁇ 15 grams) are not used. Mice are subjected to successive conditioning cycles in a hypoba ⁇ c chamber for a total of 14 days. Each 24 hour cycle consista of 18 hr at 0.40 ⁇ 0.02% atmospheric pressure and 6 hr at ambient pressure After conditioning the mice are maintained at ambient pressure for an additional 72 hr prior to dosing.
  • Test peptides or recombinant human EPO standards, are diluted in PBS + 0.1% BSA vehicle
  • PBS/BSA Peptide monomer stock solutions are first solubihzed in dimethyl sulfoxide (DMSO).
  • Negative control groups include one group of mice injected with PBS/BSA alone, and one group injected with 1% DMSO Each dose group containa 10 mice. Mice are injected subcutaneously (scruff of neck) with 0 5 mL of the appropriate sample.
  • mice Forty eight hours following sample injection, the mice are administered an intraperitoneal injection of 0.2 ml of Fe 59 (Dupont, NEN), for a dose of approximately 0.75 ⁇ Cu ⁇ es/mouse.
  • Mouse body weights are determined 24hr after Fe 59 administration, and the mice are sacrificed 48hr after Fe 59 administration. Blood is collected from each animal by cardiac puncture and hematocrits are determined (heparin was used as the anticoagulant). Each blood sample (0.2 ml) is analyzed for Fe 59 incorporation using a Packard gamma counter.
  • Non-responder mice ⁇ e , those mice with radioactive incorporation less than the negative control group) are eliminated from the appropriate data set. Mice that have hematocrit values less than 53% of the negative control group are also eliminated
  • Results are derived from sets of 10 animals for each experimental dose. The average amount of radioactivity incorporated [counts per mmute (CPM)] into blood samples from each group is calculated.
  • mice are dosed with four weekly bolus intravenous injections of either EPO positive control, test peptide, or vehicle.
  • a range of positive control and test peptide doses, expressed as mg/kg, are tested by varying the active compound concentration in the formulation. Volumes injected are 5ml/kg
  • the vehicle control group is comprised twelve animals, while 8 animals are in each of the remaining dose groups. Daily viability and weekly body weights are recorded.
  • mice are mice are fasted and then anesthetized with inhaled isoflurane and terminal blood samples are collected via cardiac or abdominal aorta puncture on Day 1 (for vehicle control mice) and on Days 15 and 29 (4 mice/group/day).
  • the blood is transferred to Vacutainer ® brand tubes.
  • Preferred anticoagulant is ethylenediaminetetraacetic acid (EDTA).
  • EDTA ethylenediaminetetraacetic acid
  • Blood samples are evaluated for endpoints measuring red blood synthesis and physiology such as hematocrit (Hct), hemoglobin (Hgb) and total erythrocyte count (RBC) using automated clinical analysers well known in the art (e g , those made by Coulter, Inc.).

Abstract

The present invention relates to peptide compounds that are agonists of the erythropoietin receptor (EPO-R). The invention further relates to therapeutic methods using such peptide compounds to treat disorders associated with insufficient or defective red blood cell production. Pharmaceutical compositions, which comprise the peptide compounds of the invention, are also provided.

Description

NOVEL PEPTIDES THAT BIND TO THE ERYTHROPOIETIN RECEPTOR
FIELD OF THE INVENTION The present invention relates to peptide compounds that are agonists of the erythropoietin receptor (EPO-R). The invention further relates to therapeutic methods using such peptide compounds to treat disorders associated with insufficient or defective red blood cell production. Pharmaceutical compositions, which comprise the peptide compounds of the invention, are also provided.
BACKGROUND OF THE INVENTION
Erythropoietin (EPO) is a glycoprotein hormone of 165 amino acids, with a molecular weight of about 34 kilodaltons (kD) and preferred glycosylation sites on amino-acid positions 24, 38, 83, and 126 It is initially produced as a precursor protein with a signal peptide of 23 amino acids. EPO can occur in three forms: α, β, and asialo The α and β forms differ slightly m their carbohydrate components, but have the same potency, biological activity, and molecular weight The asialo form is an α or β form with the terminal carbohydrate (sialic acid) removed. The DNA sequences encoding EPO have been reported [U S. Pat. No. 4,703,008 to Lm]
EPO stimulates mitotic division and differentiation of erythrocyte precursor cells, and thus ensures the production of erythrocytes It is produced in the kidney when hypoxic conditions prevail. During EPO-mduced differentiation of erythrocyte precursor cells, globm synthesis is induced; heme complex synthesis is stimulated; and the number of ferritin receptors increases. These changes allow the cell to take on more iron and synthesize functional hemoglobin, which binds in mature erythrocytes oxygen. Thus, erythrocytes and their hemoglobin play a key role in supplying the body with oxygen. These changes are initiated by the interaction of EPO with an appropriate receptor on the surface of the erythrocyte precursor cells [See, e.g., Graber and Krantz (1978) Ann. Rev. Med. 29:51-66].
EPO is present in very low concentrations in plasma when the body is in a healthy state, in which tissues receive sufficient oxygenation from the existing number of erythrocytes. This normal low EPO concentration is sufficient to stimulate replacement of red blood cells that are normally lost through aging.
The amount of EPO m the circulation is increased under conditions of hypoxia when oxygen transport by blood cells in circulation is reduced Hypoxia may be caused, for example, by substantial blood loss through hemorrhage, destruction of red blood cells by over-exposure to radiation, reduction in oxygen intake due to high altitude or prolonged unconsciousness, or vanous forms of anemia. In response to such hypoxic stress, elevated EPO levels increase red blood cell production by stimulating the proliferation of erythroid progenitor cells. When the number of red blood cells in circulation is greater than needed for normal tissue oxygen requirements, EPO levels in circulation are decreased.
Because EPO is essential in the process of red blood cell formation, this hormone has potentially useful applications in both the diagnosis and treatment of blood disorders characterized by low or defective red blood cell production. Recent studies have provided a basis for the projection of EPO therapy efficacy for a variety of disease states, disorders, and states of hematologic irregularity, including: beta-thalassemia [See Vedovato, et al. (1984) Acta. Haematol. 71:21 1-213]; cystic fibrosis [See Vichinsky, et al. (1984) J. Pediatric 105:15-21]; pregnancy and menstrual disorders [See Cotes, el al. (193) Bπt. J. Ostet. Gyneacol 90:304-311]; early anemia of prematurity [See Haga, et al. (1983) Acta Pediatr. Scand. 72; 827-831]; spinal cord injury [See Claus-Walker, et al. (1984) Arch. Phys. Med. Rehabil. 65:370-374]; space flight [See Dunn, et al. (1984) Eur. J. Appl. Physiol. 52:178-182]; acute blood loss [see, Miller, et al. (1982) Bπt. J. Haematol. 52:545-590]; aging [See Udupa, et al. (1984) J. Lab. Clin. Med. 103:574-580 and 581-588 and Lipschitz, et al. (1983) Blood 63:502-509]; various neoplastic disease states accompanied by abnormal erythropoiesis [See Dainiak, et al. (1983) Cancer 5 1101 -1 106 and Schwartz, et al. (1983) Otolaryngol. 109:269-272]; and renal insufficiency [See Eschbach. et al. (1987) N. Eng. J. Med. 316:73-78].
Purified, homogeneous EPO has been characterized [U.S. Pat. No. 4,677,195 to Hewick]. A DNA sequence encoding EPO was purified, cloned, and expressed to produce recombinant polypeptides with the same biochemical and immunological properties as natural EPO. A recombinant EPO molecule with oligosaccharides identical to those on natural EPO has also been produced [See Sasaki, et al. (1987) J. Biol. Chem. 262: 12059-12076].
The biological effect of EPO appears to be mediated, in part, by interaction with a cell membrane bound receptor. Initial studies using immature erythroid cells isolated from mouse spleen suggest that the EPO-bmding cell surface proteins comprise two polypeptides having approximate molecular weights of 85,000 Daltons and 100,000 Daltons, respectively [Sawyer, et al. (1987) Proc. Natl. Acad. Sci. USA 84:3690-3694]. The number of EPO binding sites was calculated to average from 800 to 1000 per cell surface Of these binding sites, approximately 300 bound EPO with a Kd value of approximately 90 picomolar (pM), while the remaining sites bound EPO with a reduced affinity of approximately 570 pM [Sawyer, et al. (1987) J. Biol. Chem. 262:5554-5562]. An independent study suggests that EPO-responsive splenic erythroblasts prepared from mice injected with the anemic strain (FVA) of the Friend leukemia virus possess a total of approximately 400 high and low affinity EPO binding sites with Kd values of approximately 100 pM and 800 pM, respectively [Landschulz, et al. (1989) Blood 73: 1476-1486].
Subsequent work indicated that the two forms of EPO receptor (EPO-R) were encoded by a single gene. This gene has been cloned [See, e g., Jones, et al. (1990) Blood 76, 31-35; Noguchi, et al. (1991) Blood 78:2548-2556; Maouche, et al. (1991) Blood 78:2557-2563]. For example, the DNA sequences and encoded peptide sequences for murine and human EPO-R proteins are described in PCT Pub No. WO 90/08822 to D'Andrea, et al. Current models suggest that binding of EPO to EPO- R results in the dimeπzation and activation of two EPO-R molecules, which results in subsequent steps of signal transduction [See, e g , Watowich, et al (1992) Proc. Natl. Acad. Sci. USA 89:2140- 2144]. The availability of cloned genes for EPO-R facilitates the search for agonists and antagonists of this important receptor. The availability of the recombinant receptor protein allows the study of receptor-hgand interaction in a variety of random and semi-random peptide diversity generation systems. These systems include the "peptides on plasmids" system [described in U.S. Pat. No. 6,270,170]; the "peptides on phage" system [described in U.S. Pat. No. 5,432,018 and Cwirla, et al. (1990) Proc. Natl. Acad. Sci. USA 87-6378-6382]; the "encoded synthetic library" (ESL) system [described in U.S. patent application Ser. No. 946,239, filed Sep. 16, 1992]; and the "very large scale immobilized polymer synthesis" system [described in U.S. Pat. No. 5,143,854; PCT Pub. No. 90/15070; Fodor, et al. (1991) Science 251.767-773; Dower and Fodor (1991) Ann. Rep. Med. Chem. 26-271-180; and U.S Pat. No. 5,424,186]. Peptides that interact to at least some extent with EPO-R have been identified and are described, for example, in Wrighton et al. (1996) Science 273:458-463, Johnson et al, (1998) Biochemistry 37:3699-3710, and Wrighton et al. (1997) Nat. Biotechnol. 15:1261-1265, see also U.S. Pat. Nos. 5,773,569, 5,830,851, 5,986,047, and 5,767,078; WO 96/40749; WO 96/40772; WO 01/38342; and WO 01/91780. In particular, a group of peptides containing a peptide motif has been identified, members of which bind to EPO-R and stimulate EPO-dependent cell proliferation. Yet, peptides identified to date as containing the motif stimulate EPO-dependent cell proliferation in vitro with EC50 values between about 20 nanomolar (nM) and 25OnM. Thus, peptide concentrations of 2OnM to 25OnM are required to stimulate 50% of the maximal cell proliferation stimulated by EPO Given the immense potential of EPO-R agonists, both for studies of the important biological activities mediated by this receptor and for treatment of disease, there remains a need for the identification of peptide EPO-R agonists of enhanced potency and activity. The present invention provides such compounds.
SUMMARY OF THE INVENTION
The present invention provides EPO-R agonist monomeπc peptides of dramatically enhanced potency and activity and dimeπc peptide agonists that comprise two peptide monomers. The potency of these novel peptide agonists may be further enhanced by one or more modifications, including: acetylation, intramolecular disulfide bond formation, covalent attachment of one or more polyethylene glycol (PEG) moieties, and others as listed in Figures IA - IL and throughout this application. The invention also provides peptides with protecting groups and/or hydrophobic groups. Protecting groups and/or hydrophobic groups associated with the peptides can be used to prolong half-lives of the peptides in circulation, and facilitate uptake by cells and transport across cell membranes. The invention further provides pharmaceutical compositions comprised of such peptide agonists, and methods to treat various medical conditions using such peptide agonists.
DETAILED DESCRIPTION QF THE INVENTION Brief Description of the Figure(s)
Figures IA - IL show a table of peptides, including peptide sequences of the present invention. Peptide sequences are provided using the single-letter amino acid code. Modified and non-naturally occuπng ammo acids are indicated using the abbreviations defined, infra, in this specification. For convenience, each individual peptide is referred to by reference to its unique sequence identification number (SEQ ID NO) given in the far left-hand column. Dimeπzation of individual peptides by sulfhydryl bonds ("SS bonds") is indicated m pink over the individual cysteine residues, whereas dimeπzation through the carboxyhc or amine groups (forming an amide bond) of the peptide are indicated in blue and yellow, respectively, over the involved residues. Linker moieties of the individual peptides, when present, are specified in the column labeled "Linker." The column labeled "Lmker-R" indicates the chemical moiety present as the R group, if present, on the linker.
Definitions. Unconventional amino acids in peptides are abbreviated as follows: 1 -naphthylalanine is 1-nal or Np; 2-naphthylalanine is 2-nal; N-methylglycine (also known as sarcosme) is MeG, Sc or Sar; homoseπne methylether is Hsm; and acetylated glycine (N-acetylglycine) is AcG. Other abbreviations are provided in the tables below.
As used herein, the term "polypeptide" or "protein" refers to a polymer of ammo acid monomers that are alpha ammo acids joined together through amide bonds. Polypeptides are therefore at least two amino acid residues in length, and are usually longer. Generally, the term "peptide" refers to a polypeptide that is only a few amino acid residues in length. The novel EPO-R agonist peptides of the present invention are preferably no more than about 50 amino acid residues in length. They are more preferably from about 8 to about 45 amino acid residues in length. A polypeptide, in contrast with a peptide, may comprise any number of amino acid residues. Hence, the term polypeptide included peptides as well as longer sequences of amino acids.
As used herein, the phrase "pharmaceutically acceptable" refers to molecular entities and compositions that are "generally regarded as safe," e g , that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a human. Preferably, as used herein, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U S Pharmacopeia or other generally recognized pharmacopeia for use m animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin
As used herein the term "agonist" refers to a biologically active hgand which binds to its complementary biologically active receptor and activates the latter either to cause a biological response in the receptor, or to enhance preexisting biological activity of the receptor.
The abbreviations used herein are defined in the table below and throughout the specification
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Additionally, the following are more abbreviations and their associated chemical structures.
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Nap
Figure imgf000013_0001
Nap (amide linked to N-terminus)
PEG-SPA
O
Figure imgf000014_0001
Novel peptides that are EPO-R agonists
The present invention relates to peptides that are agonists of the EPO-R and show dramatically enhanced potency and activity. These peptide agonists are preferably of about 8 to about 45 amino acids in length.
The peptides of this invention may be monomers, homo- or hetero- dimers, or other homo- or hetero- multimers. The term "homo" means comprising identical monomers; thus, for example, a homodimer of the present invention is a peptide comprising two identical monomers. The term "hetero" means comprising different monomers; thus, for example, a heterodimer of the present invention is a peptide comprising two non-identical monomers. The peptide multimers of the invention may be trimers, tetramers, pentamers, or other higher order structures. Moreover, such dimers and other multimers may be heterodimers or heteromultimers. The peptide monomers of the present invention may be degradation products (e.g., oxidation products of methionine or deamidated glutamine, arganine, and C-terminus amide). Such degradation products may be used in and are therefore considered part of the present invention. In preferred embodiments, the heteromultimers of the invention comprise multiple peptides that are all EPO-R agonist peptides. In highly preferred embodiments, the multimers of the invention are homomultimers: i.e., they comprise multiple EPO-R agonist peptides of the same amino acid sequence. Accordingly, the present invention also relates to homo- or hetero- dimeric peptide agonists of
EPO-R, which show dramatically enhanced potency and activity. In preferred embodiments, the dimers of the invention comprise two peptides that are both EPO-R agonist peptides. These preferred dimeric peptide agonists comprise two peptide monomers, wherein each peptide monomer is of about 8 to about 45 amino acids in length. In particularly preferred embodiments, the dimers of the invention comprise two EPO-R agonist peptides of the same amino acid sequence.
Stereoisomers (e.g., D-amino acids) of the twenty conventional amino acids, unnatural amino acids such as a,a-disubstituted amino acids, N-alkyl amino acids, lactic acid, and other unconventional amino acids may also be suitable components for compounds of the present invention. Examples of unconventional amino acids include, but are not limited to: β-alanine, 3-pyπdylalanine, A- hydroxyprolme, 0-phosphoseπne, N-methylglycine, N-acetylseπne, N-formylmethionine, 3- methylhistidine, 5-hydroxylysine, nor-leucine, and other similar amino acids and imino acids.
Other modifications are also possible, including modification of the amino terminus, modification of the carboxy terminus, replacement of one or more of the naturally occurring genetically encoded amino acids with an unconventional amino acid, modification of the side chain of one or more amino acid residues, peptide phosphorylation, and the like. A preferred amino terminal modification is acetylation (e g , with acetic acid or a halogen substituted acetic acid). In preferred embodiments an N-terminal glycine is acetylated to N-acetylglycine (AcG). In preferred embodiments, a the C-terminal glycine is N-methylglycme (MeG, also known as sarcosine).
In preferred embodiments, the peptide monomers of the invention contain an intramolecular disulfide bond between the two cysteine residues of the core sequence.
The present invention also provides conjugates of these peptide monomers Thus, according to a preferred embodiment, the monomeπc peptides of the present invention are dimeπzed or ohgomeπzed, thereby enhancing EPO-R agonist activity.
In one embodiment, the peptide monomers of the invention may be oligomeπzed using the biotin/streptavidin system Biotinylated analogs of peptide monomers may be synthesized by standard techniques. For example, the peptide monomers may be C-terminally biotinylated. These biotinylated monomers are then ohgomeπzed by incubation with streptavidin [e g , at a 4' 1 molar ratio at room temperature in phosphate buffered saline (PBS) or HEPES-buffered RPMI medium (Invitrogen) for 1 hour] In a variation of this embodiment, biotinylated peptide monomers may be oligomeπzed by incubation with any one of a number of commercially available anti-biotin antibodies [e g , goat anti- biotin IgG from Kirkegaard & Perry Laboratories, Inc (Washington, DC)] .
In preferred embodiments, the peptide monomers of the invention are dimeπzed by covalent attachment to at least one linker moiety. The linker (Lκ) moiety is preferably, although not necessarily, a CM2 linking moiety optionally terminated with one or two -NH- linkages and optionally substituted at one or more available carbon atoms with a lower alkyl substituent. Preferably the linker Lκ comprises -NH-R-NH- wherein R is a lower (Ci-6) alkylene substituted with a functional group such as a carboxyl group or an amino group that enables binding to another molecular moiety (e g , as may be present on the surface of a solid support). Most preferably the linker is a lysine residue or a lysine amide (a lysine residue wherein the carboxyl group has been converted to an amide moiety - CONH2). In preferred embodiments, the linker bridges the C-termini of two peptide monomers, by simultaneous attachment to the C-termmal amino acid of each monomer.
For example, when the C-terminal linker Lκ is a lysine amide the dimer may be illustrated structurally as shown in Formula I, and summarized as shown in Formula II: Formula I Formula II
Monome
Figure imgf000016_0001
In Formula I and Formula II, N2 represents the nitrogen atom of lysine's ε-ammo group and N1 represents the nitrogen atom of lysine's α-amino group. The dimeric structure can be written as [peptide]2Lys-amide to denote a peptide bound to both the α and ε amino groups of lysine, or [Ac- peptide]2Lys-amide to denote an N-terminally acetylated peptide bound to both the α and ε amino groups of lysine, or [Ac-peptide, disulfide]2Lys-amide to denote an N-termmally acetylated peptide bound to both the a and ε ammo groups of lysine with each peptide containing an intramolecular disulfide loop, or [Ac-peptide, disulfide]2Lys-spacer-PEG to denote an N-terminally acetylated peptide bound to both the α and ε ammo groups of lysine with each peptide containing an intramolecular disulfide loop and a spacer molecule forming a covalent linkage between the C- terminus of lysine and a PEG moiety, or [Ac-peptide-Lys*-NH2]2-Iminodiacetic-N-(Boc-βAla) to denote a homodimer of an Ν-termmally acetylated peptide beaπng a C-termmal lysineamide residue where the ε amine of lysine is bound to each of the two carboxyl groups of iminodiacetic acid and where Boc-beta-alamne is covalently bound to the nitrogen atom of iminodiacetic acid via an amide bond
In an additional embodiment, polyethylene glycol (PEG) may serve as the linker Lκ that dimeπzes two peptide monomers, for example, a single PEG moiety may be simultaneously attached to the Ν-termini of both peptide chains of a peptide dimer. In yet another additional embodiment, the linker (Lκ) moiety is preferably, but not necessarily, a molecule containing two carboxyhc acids and optionally substituted at one or more available atoms with an additional functional group such as an amine capable of being bound to one or more PEG molecules. Such a molecule can be depicted as.
-CO-(CH2)n-X-(CH2)m-CO- where n is an integer from 0 to 10, m is an integer from 1 to 10, X is selected from O, S, Ν(CH2)pΝRi, NCO(CH2)PNR,, and CHNR1, R, is selected from H, Boc, Cbz, etc., and p is an integer from 1 to 10.
In preferred embodiments, one amino group of each of the peptides form an amide bond with the linker Lκ. In particularly preferred embodiments, the amino group of the peptide bound to the linker Lκ is the epsilon amine of a lysine residue or the alpha amine of the N-terminal residue, or an amino group of the optional spacer molecule. In particularly preferred embodiments, both n and m are one, X is NCO(CH2)pNRi , p is two, and Ri is Boc. A dimeric EPO peptide containing such a preferred linker may be structurally illustrated as shown in Formula III.
Monomer 1 o
Figure imgf000017_0001
Formula Ul Monomer2
Optionally, the Boc group can be removed to liberate a reactive amine group capable of forming a covalent bond with a suitably activated water soluble polymer species, for example, a PEG species such as mPEG-para-mtrophenylcarbonate (mPEG-NPC), mPEG-succmimidyl propionate (mPEG- SPA), and N-hydroxysuccinimide-PEG (NHS-PEG) (see, e.g., US Patent No. 5,672,662). A dimeric EPO peptide containing such a preferred linker may be structurally illustrated as shown in Formula IV.
Monomer l
Figure imgf000017_0002
Formula IV Monomer2
Generally, although not necessarily, peptide dimers will also contain one or more intramolecular disulfide bonds between cysteine residues of the peptide monomers. Preferably, the two monomers contain at least one intramolecular disulfide bond. Most preferably, both monomers of a peptide dimer contain an intramolecular disulfide bond, such that each monomer contains a cyclic group A peptide monomer or dimer may further comprise one or more spacer moieties. Such spacer moieties may be attached to a peptide monomer or to a peptide dimer. Preferably, such spacer moieties are attached to the linker Lκ moiety that connects the monomers of a peptide dimer. For example, such spacer moieties may be attached to a peptide dimer via the carbonyl carbon of a lysine linker, or via the nitrogen atom of an iminodiacetic acid linker. For example, such a spacer may connect the linker of a peptide dimer to an attached water soluble polymer moiety or a protecting group. In another example, such a spacer may connect a peptide monomer to an attached water soluble polymer moiety. In one embodiment, the spacer moiety is a Ci.12 linking moiety optionally terminated with - NH- linkages or carboxyl (-COOH) groups, and optionally substituted at one or more available carbon atoms with a lower alkyl substituent. In one embodiment, the spacer is R-COOH wherein R is a lower (C1-I5) alkylene optionally substituted with a functional group such as a carboxyl group or an amino group that enables binding to another molecular moiety. For example, the spacer may be a glycine (G) residue, or an amino hexanoic acid. In preferred embodiments the amino hexanoic acid is 6-amino hexanoic acid (Ahx) For example, where the spacer 6-amino hexanoic acid (Ahx) is bound to the N- terminus of a peptide, the peptide terminal amine group may be linked to the carboxyl group of Ahx via a standard amide coupling. In another example, where Ahx is bound to the C-terminus of a peptide, the amine of Ahx may be linked to the carboxyl group of the linker via a standard amide coupling. The structure of such a peptide may be depicted as shown in Formula V, and summarized as shown in Formula VI.
Formula V Formula VI
Monomerl
Figure imgf000018_0001
Figure imgf000018_0002
MonomerZ
In other embodiments, the spacer is -NH-R-NH- wherein R is a lower (Q _6) alkylene substituted with a functional group such as a carboxyl group or an ammo group that enables binding to another molecular moiety. For example, the spacer may be a lysine (K) residue or a lysine amide
(K-NH2, a lysine residue wherein the carboxyl group has been converted to an amide moiety - CONH2)
In preferred embodiments, the spacer moiety has the following structure:
-NH-(CH2)α-[O-(CH2)β]γ-Oδ-(CH2)ε-Y- where α, β, γ, δ, and ε are each integers whose values are independently selected. In preferred embodiments, α, β, and ε are each integers whose values are independently selected from one to about six, δ is zero or one, γ is an integer selected from zero to about ten, except that when γ is greater than one, β is two, and Y is selected from NH or CO. In particularly preferred embodiments α, β, and ε are each equal to two, both γ and δ are equal to 1 , and Y is NH. For example, a peptide dimer containing such a spacer is illustrated schematically in Formula VII, where the linker is a lysine and the spacer joins the linker to a Boc protecting group. Formula VII
NH — Boc
Monomer L O
Monomer/ ° In another particularly preferred embodiment γ and δ are zero, α and ε together equal five, and Y is CO.
In particularly preferred embodiments, the linker plus spacer moiety has the structure shown in Formula VIII or Formula IX Formula VIII Formula IX
Figure imgf000019_0001
The peptide monomers, dimers, or multimers of the invention may further comprise one or more water soluble polymer moieties. Preferably, these polymers are covalently attached to the peptide compounds of the invention Preferably, for therapeutic use of the end-product preparation, the polymer will be pharmaceutically acceptable. One skilled m the art will be able to select the desired polymer based on such considerations as whether the polymer-peptide conjugate will be used therapeutically, and if so, the desired dosage, circulation time, resistance to proteolysis, and other considerations. The water soluble polymer may be, for example, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-l,3-dioxolane, poly-l,3,6-tπoxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), poly(n-vinyl pyrrohdone)polyethylene glycol, propropylene glycol homopolymers, polypropylene oxide/ethylene oxide copolymers, and polyoxyethylated polyols. A preferred water soluble polymer is PEG. The polymer may be of any molecular weight, and may be branched or unbranched. A preferred PEG for use m the present invention comprises linear, unbranched PEG having a molecular weight that is greater than 10 kilodaltons (kD) and is more preferably between about 20 and 60 kD in molecular weight. Still more preferably, the linear unbranched PEG moiety should have a molecular weight of between about 20 and 40 kD, with 20 kD PEG being particularly preferred. It is understood that in a given preparation of PEG, the molecular weights will typically vary among individual molecules. Some molecules will weight more, and some less, than the stated molecular weight. Such variation is generally reflect by use of the word "about" to describe molecular weights of the PEG molecules.
The number of polymer molecules attached may vary; for example, one, two, three, or more water soluble polymers may be attached to an EPO-R agonist peptide of the invention. The multiple attached polymers may be the same or different chemical moieties (e g , PEGs of different molecular weight) Thus, in a preferred embodiment the invention contemplates EPO-R agonist peptides having two or more PEG moieities attached thereto Preferably, both of the PEG moietieis are linear, unbranched PEG each preferably having a molecular weight of between about 10 and about 60 kD. more preieraoiy, each linear unbranched PEG moiety has a molecular weight that is between about 20 and 40 kD, and still more preferably between about 20 and 30 kD with a molecular weight of about 20 kD for each linear PEG moiety being particularly preferred. However, other molecular weights for PEG are also contemplated in such embodiments. For example, the invention contemplates and encompasses EPO-R agonist peptides having two or more linear unbranched PEG moieties attached thereto, at least one or both of which has a molecular weight between about 20 and 40 kD or between about 20 and 30 kD In other embodiments the invention contemplates and encompasses EPO-R agonist peptides having two or more linear unbranched PEG moieties attached thereto, at least one of which has a molecular weight between about 40 and 60 kD. In one embodiment, PEG may serve as a linker that dimeπzes two peptide monomers. In one embodiment, PEG is attached to at least one terminus (N-termmus or C-terminus) of a peptide monomer or dimer. In another embodiment, PEG is attached to a spacer moiety of a peptide monomer or dimer. In a preferred embodiment PEG is attached to the linker moiety of a peptide dimer. In a highly preferred embodiment, PEG is attached to a spacer moiety, where said spacer moiety is attached to the linker Lκ moiety that connects the monomers of a peptide dimer. Pn particularly preferred embodiments, PEG is attached to a spacer moiety, where said spacer moiety is attached to a peptide dimer via the carbonyl carbon of a lysine linker, or the amide nitrogen of a lysine amide linker.
Peptides and peptide sequences encompassed by the present invention, including peptide monomers and dimers, are shown in Figures IA - IL. For convenience, the individual peptides and peptide sequences depicted in those figures are described here by reference to Sequence Identification Numbers (SEQ ID NOs.) provided in the far left-hand column of Figures IA - IL.
The peptide sequences of the present invention can be present alone or in conjunction with N- termmal and/or C-termmal extensions of the peptide chain. Such extensions may be naturally encoded peptide sequences optionally with or substantially without non-naturally occurring sequences; the extensions may include any additions, deletions, point mutations, or other sequence modifications or combinations as desired by those skilled in the art. For example and not limitation, naturally- occurring sequences may be full-length or partial length and may include amino acid substitutions to provide a site for attachment of carbohydrate, PEG, other polymer, or the like via side chain conjugation In a variation, the amino acid substitution results in humanization of a sequence to make in compatible with the human immune system. Fusion proteins of all types are provided, including immunoglobulin sequences adjacent to or in near proximity to the EPO-R activating sequences of the present invention with or without a non-immunoglobulm spacer sequence. One type of embodiment is an immunoglobulin chain having the EPO-R activating sequence in place of the variable (V) region of the heavy and/or light chain.
Preparation of the peptide compounds of the invention: Peptide synthesis
The peptides of the invention may be prepared by classical methods known in the art. These standard methods include exclusive solid phase synthesis, partial solid phase synthesis methods, fragment condensation, classical solution synthesis, and recombinant DNA technology [See, e g , Merπfield J Am Chem Soc 1963 85-2149]
In one embodiment, the peptide monomers of a peptide dimer are synthesized individually and dimeπzed subsequent to synthesis. In preferred embodiments the peptide monomers of a dimer have the same ammo acid sequence
In particularly preferred embodiments, the peptide monomers of a dimer are linked via their C-termini by a linker Lκ moiety having two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety (e g , as may be present on the surface of a solid support). In this case, the two peptide monomers may be synthesized directly onto two reactive nitrogen groups of the linker Lκ moiety in a variation of the solid phase synthesis technique. Such synthesis may be sequential or simultaneous.
Where sequential synthesis of the peptide chains of a dimer onto a linker is to be performed, two amine functional groups on the linker molecule are protected with two different orthogonally removable amine protecting groups In preferred embodiments, the protected diamine is a protected lysine. The protected linker is coupled to a solid support via the linker's third functional group The first amine protecting group is removed, and the first peptide of the dimer is synthesized on the first deprotected amine moiety. Then the second amine protecting group is removed, and the second peptide of the dimer is synthesized on the second deprotected amine moiety. For example, the first amino moiety of the linker may be protected with Alloc, and the second with Fmoc. In this case, the Fmoc group (but not the Alloc group) may be removed by treatment with a mild base [e g , 20% pipeπdine in dimethyl formamide (DMF)], and the first peptide chain synthesized Thereafter the Alloc group may be removed with a suitable reagent [e g , Pd(PPh3)/4-methyl morpholme and chloroform], and the second peptide chain synthesized This technique may be used to generate dimers wherein the sequences of the two peptide chains are identical or different. Note that where different thiol-protectmg groups for cysteine are to be used to control disulfide bond formation (as discussed below) this technique must be used even where the final ammo acid sequences of the peptide chains of a dimer are identical.
Where simultaneous synthesis of the peptide chains of a dimer onto a linker is to be performed, two amine functional groups of the linker molecule are protected with the same removable amine protecting group In preferred embodiments, the protected diamine is a protected lysine. The protected linker is coupled to a solid support via the linker's third functional group. In this case the two protected functional groups of the linker molecule are simultaneously deprotected, and the two peptide chains simultaneously synthesized on the deprotected amines. Note that using this technique, the sequences of the peptide chains of the dimer will be identical, and the thiol-protectmg groups for the cysteine residues are all the same.
A preferred method for peptide synthesis is solid phase synthesis. Solid phase peptide synthesis procedures are well-known in the art [see, e g , Stewart Solid Phase Peptide Syntheses (Freeman and Co.: San Francisco) 1969; 2002/2003 General Catalog from Novabiochem Corp, San Diego, USA; Goodman Synthesis of Peptides and Peptidomimetics (Houben-Weyl, Stuttgart) 2002]. In solid phase synthesis, synthesis is typically commenced from the C-termmal end of the peptide using an α-amino protected resin. A suitable starting material can be prepared, for instance, by attaching the required α-amino acid to a chloromethylated resin, a hydroxymethyl resm, a polystyrene resin, a benzhydrylamine resin, or the like. One such chloromethylated resm is sold under the trade name BIO-BEADS SX-I by Bio Rad Laboratories (Richmond, CA). The preparation of the hydroxymethyl resm has been described [Bodonszky, et al. (1966) Chem. Ind. London 38: 1597] The benzhydrylamine (BHA) resin has been described [Pietta and Marshall (1970) Chem. Commun. 650], and the hydrochloride form is commercially available from Beckman Instruments, Inc. (Palo Alto, CA) For example, an α-ammo protected amino acid may be coupled to a chloromethylated resin with the aid of a cesium bicarbonate catalyst, according to the method described by Gisin (1973) HeIv. Chim Acta 56: 1467.
After initial coupling, the α-amino protecting group is removed, for example, using trifluoroacetic acid (TFA) or hydrochloric acid (HCl) solutions in organic solvents at room temperature Thereafter, α-ammo protected amino acids are successively coupled to a growing support-bound peptide chain. The α-amino protecting groups are those known to be useful in the art of stepwise synthesis of peptides, including" acyl-type protecting groups (e g , formyl, tπfluoroacetyl, acetyl), aromatic urethane-type protecting groups [e g , benzyloxycarboyl (Cbz) and substituted Cbz], aliphatic urethane protecting groups [e g , t-butyloxycarbonyl (Boc), isopropyloxycarbonyl, cyclohexyloxycarbonyl], and alkyl type protecting groups (e g , benzyl, tπphenylmethyl), fluorenylmethyl oxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), and l-(4,4-dimethyl-2,6- dioxocyclohex-l-yhdene)ethyl (Dde).
The side chain protecting groups (typically ethers, esters, tπtyl, PMC (2,2,5,7,8- pentamethyl-chroman-6-sulphonyl), and the like) remain intact during coupling and is not split off during the deprotection of the amino-terminus protecting group or during coupling. The side chain protecting group must be removable upon the completion of the synthesis of the final peptide and under reaction conditions that will not alter the target peptide. The side chain protecting groups for Tyr include tetrahydropyranyl, tert-butyl, trityl, benzyl, Cbz, Z-Br-Cbz, and 2,5-dichlorobenzyl. The side chain protecting groups for Asp include benzyl, 2,6-dichlorobenzyl, methyl, ethyl, and cyclohexyl. The side chain protecting groups for Thr and Ser include acetyl, benzoyl, trityl, tetrahydropyranyl, benzyl, 2,6-dichlorobenzyl, and Cbz The side chain protecting groups for Arg include nitro, Tosyl (Tos), Cbz, adamantyloxycarbonyl mesitoylsulfonyl (Mts), 2,2,4,6,7- pentamethyldihydrobenzofurane-5-sulfonyl (Pbf), 4-mthoxy-2,3,6-trimethyl-benzenesulfonyl (Mtr), or Boc. The side chain protecting groups for Lys include Cbz, 2-chlorobenzyloxycarbonyl (2-Cl-Cbz), 2-bromobenzyloxycarbonyl (2-Br-Cbz), Tos, or Boc After removal of the α-ammo protecting group, the remaining protected ammo acids are coupled stepwise in the desired order. Each protected amino acid is generally reacted in about a 3-fold excess using an appropriate carboxyl group activator such as 2-(lH-benzotriazol-l-yl)-l , l ,3,3 tetramethyluronium hexafluorophosphate (HBTU) or dicyclohexylcarbodimide (DCC) in solution, for example, in methylene chloride (CH2Cl2), N-methyl pyrrolidone, dimethyl formamide (DMF), or mixtures thereof.
After the desired amino acid sequence has been completed, the desired peptide is decoupled from the resin support by treatment with a reagent, such as tπfiuoroacetic acid (TFA) or hydrogen fluoride (HF), which not only cleaves the peptide from the resin, but also cleaves all remaining side chain protecting groups When a chloromethylated resin is used, hydrogen fluoride treatment results in the formation of the free peptide acids. When the benzhydrylamine resm is used, hydrogen fluoride treatment results directly m the free peptide amide Alternatively, when the chloromethylated resin is employed, the side chain protected peptide can be decoupled by treatment of the peptide resin with ammonia to give the desired side chain protected amide or with an alkylamine to give a side chain protected alkylamide or dialkylamide. Side chain protection is then removed in the usual fashion by treatment with hydrogen fluoride to give the free amides, alkylamides, or dialkylamides In prepaπng the esters of the invention, the resins used to prepare the peptide acids are employed, and the side chain protected peptide is cleaved with base and the appropriate alcohol (e g , methanol) Side chain protecting groups are then removed in the usual fashion by treatment with hydrogen fluoride to obtain the desired ester. These procedures can also be used to synthesize peptides in which ammo acids other than the
20 naturally occurring, genetically encoded amino acids are substituted at one, two, or more positions of any of the compounds of the invention. Synthetic ammo acids that can be substituted into the peptides of the present invention include, but are not limited to, N-methyl, L-hydroxypropyl, L-3, A- dihydroxyphenylalanyl, δ amino acids such as L- δ-hydroxylysyl and D- δ-methylalanyl, L-α- methylalanyl, β amino acids, and isoquinolyl D-amino acids and non-naturally occurring synthetic amino acids can also be incorporated into the peptides of the present invention.
Peptide modifications
One can also modify the amino and/or carboxy termini of the peptide compounds of the invention to produce other compounds of the invention. Ammo terminus modifications include methylation (e g , -NHCH3 or ~N(CH3)2), acetylation (e g , with acetic acid or a halogenated derivative thereof such as α-chloroacetic acid, α-bromoacetic acid, or α-iodoacetic acid), adding a benzyloxycarbonyl (Cbz) group, or blocking the ammo terminus with any blocking group containing a carboxylate functionality defined by RCOO-- or sulfonyl functionality defined by R-SO2 --, where R is selected from alkyl, aryl, heteroaryl, alkyl aryl, and the like, and similar groups One can also incoφorate a desamino acid at the N-termmus (so that there is no N-termmal amino group) to decrease susceptibility to proteases or to restrict the conformation of the peptide compound In preferred embodiments, the N-terminus is acetylated In particularly preferred embodiments an N- terminal glycine is acetylated to yield N-acetylglycme (AcG)
Carboxy terminus modifications include replacing the free acid with a carboxamide group or forming a cyclic lactam at the carboxy terminus to introduce structural constraints One can also cychze the peptides of the invention, or incorporate a desammo or descarboxy residue at the termini of the peptide, so that there is no terminal amino or carboxyl group, to decrease susceptibility to proteases or to restrict the conformation of the peptide C-terminal functional groups of the compounds of the present invention include amide, amide lower alkyl, amide di(lower alkyl), lower alkoxy, hydroxy, and carboxy, and the lower ester derivatives thereof, and the pharmaceutically acceptable salts thereof
One can replace the naturally occurring side chains of the 20 genetically encoded ammo acids (or the stereoisomeπc D amino acids) with other side chains, for instance with groups such as alkyl, lower alkyl, cyclic 4-, 5-, 6-, to 7-membered alkyl, amide, amide lower alkyl, amide di(lower alkyl), lower alkoxy, hydroxy, carboxy and the lower ester derivatives thereof, and with 4-, 5-, 6-, to 7- membered heterocyclic In particular, proline analogues in which the ring size of the proline residue is changed from 5 members to 4, 6, or 7 members can be employed Cyclic groups can be saturated or unsaturated, and if unsaturated, can be aromatic or non-aromatic Heterocyclic groups preferably contain one or more nitrogen, oxygen, and/or sulfur heteroatoms Examples of such groups include the furazanyl, furyl, lmidazohdinyl, lmidazolyl, lmidazohnyl, isothiazolyl, isoxazolyl, morpholinyl {e g morphohno), oxazolyl, prperazmyl {e g , 1 -piperazmyl), pipeπdyl {e g , 1-pipeπdyl, pipeπdmo), pyranyl, pyrazinyl, pyrazohdmyl, pyrazolmyl, pyrazolyl, pyπdazinyl, pyridyl, pyrimidinyl, pyrrolidinyl {e g , 1-pyrrolidinyl), pyrrohnyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, thiomorphohnyl {e g , thiomorpholino), and triazolyl These heterocyclic groups can be substituted or unsubstituted Where a group is substituted, the substituent can be alkyl, alkoxy, halogen, oxygen, or substituted or unsubstituted phenyl
One can also readily modify peptides by phosphorylation, and other methods [e g , as described in Hruby, et al (1990) Biochem J 268 249-262]
The peptide compounds of the invention also serve as structural models for non-peptidic compounds with similar biological activity Those of skill in the art recognize that a variety of techniques are available for constructing compounds with the same or similar desired biological activity as the lead peptide compound, but with more favorable activity than the lead with respect to solubility, stability, and susceptibility to hydrolysis and proteolysis [See, Morgan and Gamor (1989) Ann Rep. Med Chem. 24:243-252]. These techniques include replacing the peptide backbone with a backbone composed of phosphonates, amidates, carbamates, sulfonamides, secondary amines, and N- methylamino acids.
Formation of disulfide bonds
The compounds of the present invention may contain one or more intramolecular disulfide bonds. In one embodiment, a peptide monomer or dimer comprises at least one intramolecular disulfide bond. In preferred embodiments, a peptide dimer comprises two intramolecular disulfide bonds.
Such disulfide bonds may be formed by oxidation of the cysteine residues of the peptide core sequence. In one embodiment the control of cysteine bond formation is exercised by choosing an oxidizing agent of the type and concentration effective to optimize formation of the desired isomer. For example, oxidation of a peptide dimer to form two intramolecular disulfide bonds (one on each peptide chain) is preferentially achieved (over formation of intermolecular disulfide bonds) when the oxidizing agent is DMSO.
In preferred embodiments, the formation of cysteine bonds is controlled by the selective use of thiol-protecting groups during peptide synthesis. For example, where a dimer with two intramolecular disulfide bonds is desired, the first monomer peptide chain is synthesized with the two cysteine residues of the core sequence protected with a first thiol protecting group [e g , trityl(Trt), allyloxycarbonyl (Alloc), and l-(4,4-dimethyl-2,6-dioxocyclohex-l-yhdene)ethyl (Dde) or the like], then the second monomer peptide is synthesized the two cysteine residues of the core sequence protected with a second thiol protecting group different from the first thiol protecting group [e g , acetamidomethyl (Acm), t-butyl (tBu), or the like]. Thereafter, the first thiol protecting groups are removed effecting bisulfide cychzation of the first monomer, and then the second thiol protecting groups are removed effecting bisulfide cychzation of the second monomer.
Other embodiments of this invention provide for analogues of these disulfide derivatives in which one of the sulfurs has been replaced by a CH2 group or other isotere for sulfur. These analogues can be prepared from the compounds of the present invention, wherein each core sequence contains at least one C or homocysteine residue and an α-amino-γ-butyπc acid in place of the second C residue, via an intramolecular or intermolecular displacement, using methods known m the art [See, e g , Barker, et al. (1992) J. Med. Chem. 35:2040-2048 and Or, et al. (1991) J. Org. Chem. 56:3146-3149] One of skill in the art will readily appreciate that this displacement can also occur using other homologs of α-amino-γ-butyπc acid and homocysteine. In addition to the foregoing cychzation strategies, other non-disulfide peptide cychzation strategies can be employed. Such alternative cychzation strategies include, for example, amide- cychzation strategies as well as those involving the formation of thio-ether bonds. Thus, the compounds of the present invention can exist in a cyclized form with either an intramolecular amide bond or an intramolecular thio-ether bond. For example, a peptide may be synthesized wherein one cysteine of the core sequence is replaced with lysine and the second cysteine is replaced with glutamic acid Thereafter a cyclic monomer may be formed through an amide bond between the side chains of these two residues Alternatively, a peptide may be synthesized wherein one cysteine of the core sequence is replaced with lysine. A cyclic monomer may then be formed through a thio-ether linkage between the side chains of the lysine residue and the second cysteine residue of the core sequence As such, in addition to disulfide cychzation strategies, amide-cychzation strategies and thio-ether cychzation strategies can both be readily used to cyclize the compounds of the present invention. Alternatively, the amino-terminus of the peptide can be capped with an α-substituted acetic acid, wherein the α-substituent is a leaving group, such as an α-haloacetic acid, for example, α - chloroacetic acid, α -bromoacetic acid, or α-iodoacetic acid.
Addition of linkers
In embodiments where a peptide dimer is dimeπzed by a linker Lκ moiety, said linker may be incorporated into the peptide during peptide synthesis. For example, where a linker Lκ moiety contains two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety, the linker may be conjugated to a solid support Thereafter, two peptide monomers may be synthesized directly onto the two reactive nitrogen groups of the linker Lκ moiety in a variation of the solid phase synthesis technique
In alternate embodiments where a peptide dimer is dimeπzed by a linker Lκ moiety, said linker may be conjugated to the two peptide monomers of a peptide dimer after peptide synthesis Such conjugation may be achieved by methods well established in the art. In one embodiment, the linker contains at least two functional groups suitable for attachment to the target functional groups of the synthesized peptide monomers For example, a linker with two free amine groups may be reacted with the C-terminal carboxyl groups of each of two peptide monomers. In another example, linkers containing two carboxyl groups, either preactivated or in the presence of a suitable coupling reagent, may be reacted with the N-terminal or side chain amine groups, or C-termmal lysine amides, of each of two peptide monomers
Addition of spacers
In embodiments where the peptide compounds contain a spacer moiety, said spacer may be incorporated into the peptide during peptide synthesis. For example, where a spacer contains a free amino group and a second functional group (e g , a carboxyl group or an ammo group) that enables binding to another molecular moiety, the spacer may be conjugated to the solid support. Thereafter, the peptide may be synthesized directly onto the spacer's free amino group by standard solid phase techniques.
In a preferred embodiment, a spacer containing two functional groups is first coupled to the solid support via a first functional group. Next a linker Lκ moiety having two functional groups capable of serving as initiation sites for peptide synthesis and a third functional group (e g , a carboxyl group or an amino group) that enables binding to another molecular moiety is conjugated to the spacer via the spacer's second functional group and the linker's third functional group Thereafter, two peptide monomers may be synthesized directly onto the two reactive nitrogen groups of the linker Lκ moiety in a variation of the solid phase synthesis technique. For example, a solid support coupled spacer with a free amine group may be reacted with a lysine linker via the linker's free carboxyl group.
In alternate embodiments where the peptide compounds contain a spacer moiety, said spacer may be conjugated to the peptide after peptide synthesis Such conjugation may be achieved by methods well established in the art In one embodiment, the linker contains at least one functional group suitable for attachment to the target functional group of the synthesized peptide. For example, a spacer with a free amine group may be reacted with a peptide's C-terminal carboxyl group. In another example, a linker with a free carboxyl group may be reacted with the free amine group of a peptide's N-terminus or of a lysine residue. In yet another example, a spacer containing a free sulfhydryl group may be conjugated to a cysteine residue of a peptide by oxidation to form a disulfide bond
Attachment of water soluble polymers
Included with the below description, the U.S. Patent Application Serial Number 10/844,933 and International Patent Application No. PCT/US04/14887, filed May 12, 2004, are incorporated by reference herein in their entirety.
In recent years, water-soluble polymers, such as polyethylene glycol (PEG), have been used for the covalent modification of peptides of therapeutic and diagnostic importance Attachment of such polymers is thought to enhance biological activity, prolong blood circulation time, reduce lmmunogenicity, increase aqueous solubility, and enhance resistance to protease digestion. For example, covalent attachment of PEG to therapeutic polypeptides such as mterleukins [Knauf, et al.
(1988) J. Biol. Chem. 263;15064; Tsutsumi, et al. (1995) J. Controlled Release 33:447), interferons
(Kita, et al (1990) Drug Des. Delivery 6:157), catalase (Abuchowski, et al. (1977) J. Biol. Chem.
252.582), superoxide dismutase (Beauchamp, et al. (1983) Anal. Biochem. 131 '25), and adenosine deaminase (Chen, et al. (1981) Biochim. Biophy. Acta 660:293), has been reported to extend their half life in vivo, and/or reduce their lmmunogenicity and antigenicity The peptide compounds of the invention may further comprise one or more water soluble polymer moieties Preferably, these polymers are covalently attached to the peptide compounds. The water soluble polymer may be, for example, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrohdone, poly-l,3-dioxolane, poly-l,3,6-tπoxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, polypropylene oxide/ethylene oxide copolymers, and polyoxyethylated polyols. A preferred water soluble polymer is PEG.
Peptides, peptide dimers and other peptide -based molecules of the invention can be attached to water-soluble polymers (e.g., PEG) using any of a variety of chemistries to link the water-soluble polymer(s) to the receptor-binding portion of the molecule (e.g , peptide + spacer). A typical embodiment employs a single attachment junction for covalent attachment of the water soluble polymer(s) to the receptor-binding portion, however in alternative embodiments multiple attachment junctions may be used, including further variations wherein different species of water-soluble polymer are attached to the receptor-bindmg portion at distinct attachment junctions, which may include covalent attachment junction(s) to the spacer and/or to one or both peptide chains. In some embodiments, the dimer or higher order multimer will comprise distinct species of peptide chain (i e., a heterodimer or other heteromul timer). By way of example and not limitation, a dimer may comprise a first peptide chain having a PEG attachment junction and the second peptide chain may either lack a PEG attachment junction or utilize a different linkage chemistry than the first peptide chain and in some variations the spacer may contain or lack a PEG attachment junction and said spacer, if PEGylated, may utilize a linkage chemistry different than that of the first and/or second peptide chains. An alternative embodiment employs a PEG attached to the spacer portion of the receptor- binding portion and a different water-soluble polymer (e.g., a carbohydrate) conjugated to a side chain of one of the amino acids of the peptide portion of the molecule.
A wide variety of polyethylene glycol (PEG) species may be used for PEGylation of the receptor-binding portion (peptides + spacer). Substantially any suitable reactive PEG reagent can be used In preferred embodiments, the reactive PEG reagent will result in formation of a carbamate or amide bond upon conjugation to the receptor-binding portion. Suitable reactive PEG species include, but are not limited to, those which are available for sale in the Drug Delivery Systems catalog (2003) of NOF Corporation (Yebisu Garden Place Tower, 20-3 Ebisu 4-chome, Shibuya-ku, Tokyo 150- 6019) and the Molecular Engineering catalog (2003) of Nektar Therapeutics (490 Discovery Drive, Huntsville, Alabama 35806) For example and not limitation, the following PEG reagents are often preferred in various embodiments: mPEG2-NHS, mPEG2-ALD, multi-Arm PEG, mPEG(MAL)2, mPEG2(MAL), mPEG-NH2, mPEG-SPA, mPEG-SBA, mPEG-thioesters, mPEG-Double Esters, mPEG-BTC, mPEG-ButyrALD, mPEG-ACET, heterofunctional PEGs (NH2-PEG-COOH, Boc- PEG-NHS, Fmoc-PEG-NHS, NHS-PEG-VS, NHS-PEG-MAL), PEG acrylates (ACRL-PEG-NHS), PEG-phosphohpids (e.g., mPEG-DSPE), multiarmed PEGs of the SUNBRITE series including the GL series of glyceπne-based PEGs activated by a chemistry chosen by those skilled in the art, any of the SUNBRITE activated PEGs (including but not limited to carboxyl-PEGs, p-NP-PEGs, Tresyl- PEGs, aldehyde PEGs, acetal-PEGs, amino-PEGs, thiol-PEGs, maleimido-PEGs, hydroxyl-PEG- amine, amino-PEG-COOH, hydroxyl-PEG-aldehyde, carboxylic anhydride type-PEG, functionalized PEG-phosphohpid, and other similar and/or suitable reactive PEGs as selected by those skilled in the art for their particular application and usage.
The polymer may be of any molecular weight, and may be branched or unbranched. A preferred PEG for use in the present invention comprises linear, unbranched PEG having a molecular weight of from about 20 kilodaltons (kD or kDa) to about 4OkD (the term "about" indicating that in preparations of PEG, some molecules will weigh more, some less, than the stated molecular weight). Most preferably, the PEG has a molecular weight of from about 3OkD to about 4OkD. Other sizes may be used, depending on the desired therapeutic profile (e g , duration of sustained release desired; effects, if any, on biological activity; ease in handling; degree or lack of antigenicity; and other known effects of PEG on a therapeutic peptide).
The number of polymer molecules attached may vary; for example, one, two, three, or more water soluble polymers may be attached to an EPO-R agonist peptide of the invention. The multiple attached polymers may be the same or different chemical moieties (e g , PEGs of different molecular weight) In some cases, the degree of polymer attachment (the number of polymer moieties attached to a peptide and/or the total number of peptides to which a polymer is attached) may be influenced by the proportion of polymer molecules versus peptide molecules in an attachment reaction, as well as by the total concentration of each in the reaction mixture. In general, the optimum polymer versus peptide ratio (in terms of reaction efficiency to provide for no excess unreacted peptides and/or polymer moieties) will be determined by factors such as the desired degree of polymer attachment (e g , mono, di-, tri-, etc.), the molecular weight of the polymer selected, whether the polymer is branched or unbranched, and the reaction conditions for a particular attachment method.
In preferred embodiments, the covalently attached water soluble polymer is PEG. For illustrative purposes, examples of methods for covalent attachment of PEG (PEGylation) are described below. These illustrative descriptions are not intended to be limiting. One of ordinary skill in the art will appreciate that a variety of methods for covalent attachment of a broad range of water soluble polymers is well established in the art. As such, peptide compounds to which any of a number of water soluble polymers known in the art have been attached by any of a number of attachment methods known in the art are encompassed by the present invention.
In one embodiment, PEG may serve as a linker that dimeπzes two peptide monomers. In one embodiment, PEG is attached to at least one terminus (N-termmus or C-terminus) of a peptide monomer or dimer. In another embodiment, PEG is attached to a spacer moiety of a peptide monomer or dimer. In a preferred embodiment PEG is attached to the linker moiety of a peptide dimer. In a highly preferred embodiment, PEG is attached to a spacer moiety, where said spacer moiety is attached to the linker Lκ moiety that connects the monomers of a peptide dimer. Most preferably, PEG is attached to a spacer moiety, where said spacer moiety is attached to a peptide dimer via the carbonyl carbon of a lysine linker, or the amide nitrogen of a lysine amide linker. There are a number of PEG attachment methods available to those skilled in the art [see, e g ,
Goodson, et al. (1990) Bio/Technology 8.343 (PEGylation of interleukm-2 at its glycosylation site after site-directed mutagenesis); EP 0 401 384 (coupling PEG to G-CSF); Malik, et al., (1992) Exp. Hematol. 20: 1028-1035 (PEGylation of GM-CSF using tresyl chloride); PCT Pub. No. WO 90/12874 (PEGylation of erythropoietin containing a recombinantly introduced cysteine residue using a cysteine-specific mPEG derivative); U.S. Pat. No 5,757,078 (PEGylation of EPO peptides), and U.S Pat No. 6,077,939 (PEGylation of an N-terminal α-carbon of a peptide)]
For example, PEG may be covalently bound to ammo acid residues via a reactive group. Reactive groups are those to which an activated PEG molecule may be bound (e g , a free amino or carboxyl group). For example, N-terminal amino acid residues and lysine (K) residues have a free amino group; and C-terminal ammo acid residues have a free carboxyl group. Sulfhydryl groups (e g , as found on cysteine residues) may also be used as a reactive group for attaching PEG. In addition, enzyme -assisted methods for introducing activated groups (e g , hydrazide, aldehyde, and aromatic- ammo groups) specifically at the C-terminus of a polypeptide have been described [Schwarz, et al. (1990) Methods Enzymol. 184:160, Rose, et al. (1991) Bioconjugate Chem 2:154; Gaertner, et al. (1994) J. Biol. Chem. 269:7224].
For example, PEG molecules may be attached to peptide amino groups using methoxylated PEG ("mPEG") having different reactive moieties. Such polymers include mPEG-succinimidyl succinate, mPEG-succmimidyl carbonate, mPEG-imidate, mPEG-4-nitrophenyl carbonate, and mPEG-cyanuπc chloride. Similarly, PEG molecules may be attached to peptide carboxyl groups using methoxylated PEG with a free amine group (mPEG-NH2)
Where attachment of the PEG is non-specific and a peptide containing a specific PEG attachment is desired, the desired PEGylated compound may be purified from the mixture of PEGylated compounds. For example, if an N-terminally PEGylated peptide is desired, the N-terminally PEGylated form may be purified from a population of randomly PEGylated peptides (ι e , separating this moiety from other monoPEGylated moieties).
In preferred embodiments, PEG is attached site-specifically to a peptide. Site-specific PEGylation at the N-terminus, side chain, and C-terminus of a potent analog of growth hormone- releasing factor has been performed through solid-phase synthesis [Felix, et al. (1995) Int. J. Peptide Protein Res 46'253]. Another site-specific method involves attaching a peptide to extremities of liposomal surface-grafted PEG chains in a site-specific manner through a reactive aldehyde group at the N-terminus generated by sodium peπodate oxidation of N-terminal threonine [Zahpsky, et al. (1995) Bioconj Chem. 6:705], However, this method is limited to polypeptides with N-terminal serine or threonine residues. Another site-specific method for N-terminal PEGylation of a peptide via a hydrazone, reduced hydrazone, oxime, or reduced oxime bond is described in U.S. Pat. No. 6,077,939 to Wei, et al.
In one method, selective N-termmal PEGylation may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for deπvatization in a particular protein. Under the appropriate reaction conditions, a carbonyl group containing PEG is selective attached to the N-termmus of a peptide. For example, one may selectively N-terminally PEGylate the protein by performing the reaction at a pH which exploits the pKa differences between the ε-amino groups of a lysine residue and the α- amino group of the N-termmal residue of the peptide By such selective attachment, PEGylation takes place predominantly at the N-termmus of the protein, with no significant modification of other reactive groups {e g , lysine side chain amino groups). Using reductive alkylation, the PEG should have a single reactive aldehyde for coupling to the protein (e g , PEG propπonaldehyde may be used).
Site-specific mutagenesis is a further approach which may be used to prepare peptides for site-specific polymer attachment. By this method, the ammo acid sequence of a peptide is designed to incorporate an appropriate reactive group at the desired position within the peptide. For example, WO 90/12874 describes the site-directed PEGylation of proteins modified by the insertion of cysteine residues or the substitution of other residues for cysteine residues. This publication also describes the preparation of mPEG-erythropoietin ("mPEG-EPO") by reacting a cysteme-specific mPEG derivative with a recombinantly introduced cysteine residue on EPO.
Where PEG is attached to a spacer or linker moiety, similar attachment methods may be used. In this case, the linker or spacer contains a reactive group and an activated PEG molecule containing the appropπate complementary reactive group is used to effect covalent attachment. In preferred embodiments the linker or spacer reactive group contains a terminal amino group (/ e , positioned at the terminus of the linker or spacer) which is reacted with a suitably activated PEG molecule to make a stable covalent bond such as an amide or a carbamate. Suitable activated PEG species include, but are not limited to, mPEG-para-nitrophenylcarbonate (mPEG-NPC), mPEG-succinimidyl carbonate (mPEG-SC), and mPEG-succinimidyl propionate (mPEG-SPA). In other preferred embodiments, the linker or spacer reactive group contains a carboxyl group capable of being activated to form a covalent bond with an amine-containmg PEG molecule under suitable reaction conditions. Suitable PEG molecules include mPEG-NB^ and suitable reaction conditions include carbodnmide-mediated amide formation or the like.
EPO-R agonist activity assays.
The biological activity of the various peptide compounds of this invention (e.g., as EPO-R agonists) can be assayed by any of a variety of methods that are well known in the art. See, for example, in International Patent Application No. PCT/US04/14886, filed May 12, 2004 Non-hmiting examples of certain, preferred assays are also described here.
In vitro functional assays In vitro competitive binding assays quantitate the ability of a test peptide to compete with
EPO for binding to EPO-R For example (see, e g , as described in U.S. Patent 5,773,569), the extracellular domain of the human EPO-R (EPO binding protein, EBP) may be recombinantly produced in E coll and the recombinant protein coupled to a solid support, such as a microtitre dish or a synthetic bead [e g , Sulfolink beads from Pierce Chemical Co. (Rockford, IL)]. Immobilized EBP is then incubated with labeled recombinant EPO, or with labeled recombinant EPO and a test peptide Serial dilutions of test peptide are employed for such experiments. Assay points with no added test peptide define total EPO binding to EBP. For reactions containing test peptide, the amount of bound EPO is quantitated and expressed as a percentage of the control (total=100%) binding. These values are plotted versus peptide concentration The IC50 value is defined as the concentration of test peptide which reduces the binding of EPO to EBP by 50% (ι e , 50% inhibition of EPO binding).
A different in vitro competitive binding assay measures the light signal generated as a function of the proximity of two beads: an EPO-conjugated bead and an EPO-R-conjugated bead. Bead proximity is generated by the binding of EPO to EPO-R. A test peptide that competes with EPO for binding to EPO-R will prevent this binding, causing a decrease in light emission The concentration of test peptide that results in a 50% decrease in light emission is defined as the IC50 value
The biological activity and potency of monomeric and dimeric peptide EPO-R agonists of the invention, which bind specifically to the EPO-receptor, may be measured using in vitro cell-based functional assays One assay is based upon a murine pre-B-cell line expressing human EPO-R and further transfected with a fos promoter-driven luciferase reporter gene construct. Upon exposure to EPO or another EPO-R agonist, such cells respond by synthesizing luciferase Luciferase causes the emission of light upon addition of its substrate lucifeπn Thus, the level of EPO-R activation in such cells may be quantitated via measurement of luciferase activity The activity of a test peptide is measured by adding serial dilutions of the test peptide to the cells, which are then incubated for 4 hours. After incubation, lucifeπn substrate is added to the cells, and light emission is measured. The concentration of test peptide that results in a half-maximal emission of light is recorded as the EC50
Another assay may be performed using FDC-P1/ER cells [Dexter, et al. (1980) J. Exp Med 152 1036-1047], a well characterized nontransformed murine bone marrow derived cell line into which EPO-R has been stably transfected These cells exhibit EPO-dependent proliferation.
In one such assay, the cells are grown to half stationary density in the presence of the necessary growth factors (see, e g , as described in U.S. Patent 5,773,569). The cells are then washed in PBS and starved for 16-24 hours in whole media without the growth factors. After determining the viability of the cells (eg , by trypan blue staining), stock solutions (in whole media without the growth factors) are made to give about 105 cells per 50μL Serial dilutions of the peptide EPO-R agonist compounds (typically the free, solution phase peptide as opposed to a phage-bound or other bound or immobilized peptide) to be tested are made in 96-well tissue culture plates for a final volume of 50μL per well Cells (50μL) are added to each well and the cells are incubated 24-48 hours, at which point the negative controls should die or be quiescent. Cell proliferation is then measured by techniques known m the art, such as an MTT assay which measures H3-thymidme incorporation as an indication of cell proliferation [see, Mosmann (1983) J Immunol Methods 65-55-63]. Peptides are evaluated on both the EPO-R-expressing cell line and a parental non-expressing cell line. The concentration of test peptide necessary to yield one half of the maximal cell proliferation is recorded as the EC50.
Li another assay, the cells are grown to stationary phase m EPO-supplemented medium, collected, and then cultured for an additional 18 hr in medium without EPO. The cells are divided into three groups of equal cell density: one group with no added factor (negative control), a group with EPO (positive control), and an experimental group with the test peptide. The cultured cells are then collected at various time points, fixed, and stained with a DNA-binding fluorescent dye (e g , propidium iodide or Hoechst dye, both available from Sigma). Fluorescence is then measured, for example, using a FACS Scan Flow cytometer. The percentage of cells m each phase of the cell cycle may then be determined, for example, using the SOBR model of CeIIFIT software (Becton Dickinson). Cells treated with EPO or an active peptide will show a greater proportion of cells in S phase (as determined by increased fluorescence as an indicator of increased DNA content) relative to the negative control group.
Similar assays may be performed using FDCP-I [see, e g , Dexter et al. (1980) J. Exp. Med. 152.1036-1047] or TF-I [Kitamura, et al. (1989) Blood 73-375-380] cell lines. FDCP-I is a growth factor dependent murine multi-potential primitive hematopoietic progenitor cell line that can proliferate, but not differentiate, when supplemented with WEHI-3 -conditioned media (a medium that contains IL-3, ATCC number TEB-68). For such experiments, the FDCP-I cell line is transfected with the human or murine EPO-R to produce FDCP- 1-hEPO-R or FDCP- 1 -mEPO-R cell lines, respectively, that can proliferate, but not differentiate, in the presence of EPO. TF-I, an EPO- dependent cell line, may also be used to measure the effects of peptide EPO-R agonists on cellular proliferation.
In yet another assay, the procedure set forth m Krystal (1983) Exp. Hematol 1 1:649-660 for a microassay based on H3-thymidine incorporation into spleen cells may be employed to ascertain the ability of the compounds of the present invention to serve as EPO agonists. In brief, B6C3F, mice are injected daily for two days with phenylhydrazine (60 mg/kg). On the third day, spleen cells are removed and their ability to proliferate over a 24 hour period ascertained using an MTT assay. The binding of EPO to EPO-R in an erythropoietin-responsive cell line induces tyrosine phosphorylation of both the receptor and numerous intracellular proteins, including She, vav and JAK2 kinase. Therefore, another in vitro assay measures the ability of peptides of the invention to induce tyrosine phosphorylation of EPO-R and downstream intracellular signal transducer proteins. Active peptides, as identified by binding and proliferation assays described above, elicit a phosphorylation pattern nearly identical to that of EPO in erythropoietin-responsive cells. For this assay, FDC-P1/ER cells [Dexter, et al. (1980) J Exp Med 152- 1036-47] are maintained in EPO- supplemented medium and grown to stationary phase. These cells are then cultured in medium without EPO for 24 hr. A defined number of such cells is then incubated with a test peptide for approximately 10 min at 37°C A control sample of cells with EPO is also run with each assay. The treated cells are then collected by centrifugation, resuspended in SDS lysis buffer, and subjected to SDS polyacrylamide gel electrophoresis. The electrophoresed proteins in the gel are transferred to nitrocellulose, and the phosphotyrosine containing proteins on the blot visualized by standard immunological techniques. For example, the blot may be probed with an anti-phosphotyrosme antibody (e g , mouse anti -phosphotyrosine IgG from Upstate Biotechnology, Inc.), washed, and then probed with a secondary antibody [e g , peroxidase labeled goat anti-mouse IgG from Kirkegaard & Perry Laboratories, Inc. (Washington, DC)]. Thereafter, phosphotyrosine-containing proteins may be visualized by standard techniques including coloπmetπc, chemiluminescent, or fluorescent assays. For example, a chemiluminescent assay may be performed using the ECL Western Blotting System from Amersham
Another cell-based in vitro assay that may be used to assess the activity of the peptides of the present invention comprises a colony assay, using murine bone marrow or human peripheral blood cells. Murine bone marrow may be obtained from the femurs of mice, while a sample of human peripheral blood may obtained from a healthy donor. In the case of peripheral blood, mononuclear cells are first isolated from the blood, for example, by centrifugation through a Ficoll-Hypaque gradient [Stem Cell Technologies, Inc. (Vancouver, Canada)]. For this assay a nucleated cell count is performed to establish the number and concentration of nucleated cells in the original sample. A defined number of cells is plated on methyl cellulose as per manufacturer's instructions [Stem Cell Technologies, Inc. (Vancouver, Canada)]. An experimental group is treated with a test peptide, a positive control group is treated with EPO, and a negative control group receives no treatment. The number of growing colonies for each group is then scored after defined periods of incubation, generally 10 days and 18 days. An active peptide will promote colony formation.
Other in vitro biological assays that can be used to demonstrate the activity of the compounds of the present invention are disclosed in Greenberger, et al. (1983) Proc. Natl. Acad. Sci. USA 80-2931-2935 (EPO-dependent hematopoietic progenitor cell line); Quelle and Wojchowski (1991) J. Biol. Chem 266:609-614 (protein tyrosine phosphorylation in BόSUt.EP cells); Dusanter-Fourt, et al. (1992) J. Biol. Chem. 287.10670-10678 (tyrosine phosphorylation of EPO-receptor in human EPO- responsive cells); Quelle, et al. (1992) J. Biol Chem 267.17055-17060 (tyrosine phosphorylation of a cytosohc protein, pp 100, in FDC-ER cells); Worthington, et al. (1987) Exp. Hematol. 15:85-92 (coloπmetric assay for hemoglobin); Kaiho and Miuno (1985) Anal. Biochem. 149: 117-120 (detection of hemoglobin with 2,7-diaminofluorene); Patel, et al. (1992) J. Biol. Chem. 267:21300- 21302 (expression of c-myb); Witthuhn, et al. (1993) Cell 74:227-236 (association and tyrosine phosphorylation of JAK2); Leonard, et al. (1993) Blood 82: 1071-1079 (expression of GATA transcription factors); and Ando, et al. (1993) Proc. Natl. Acad. Sci. USA 90:9571-9575 (regulation of Gi transition by cycling D2 and D3).
An instrument designed by Molecular Devices Corp., known as a microphysiometer, has been reported to be successfully used for measurement of the effect of agonists and antagonists on various receptors. The basis for this apparatus is the measurement of the alterations in the acidification rate of the extracellular media in response to receptor activation.
In vivo functional assays One in vivo functional assay that may be used to assess the potency of a test peptide is the polycythemic exhypoxic mouse bioassay. For this assay, mice are subjected to an alternating conditioning cycle for several days. In this cycle, the mice alternate between periods of hypobaric conditions and ambient pressure conditions. Thereafter, the mice are maintained at ambient pressure for 2-3 days prior to administration of test samples Test peptide samples, or EPO standard m the case positive control mice, are injected subcutaneously into the conditioned mice. Radiolabeled iron (e g , Fe59) is administered 2 days later, and blood samples taken two days after administration of radiolabeled iron. Hematocrits and radioactivity measurements are then determined for each blood sample by standard techniques Blood samples from mice injected with active test peptides will show greater radioactivity (due to binding of Fe59 by erythrocyte hemoglobin) than mice that did not receive test peptides or EPO.
Another in vivo functional assay that may be used to assess the potency of a test peptide is the reticulocyte assay. For this assay, normal untreated mice are subcutaneously injected on three consecutive days with either EPO or test peptide. On the third day, the mice are also intraperitoneally injected with iron dextran. At day five, blood samples are collected from the mice. The percent (%) of reticulocytes in the blood is determined by thiazole orange staining and flow cytometer analysis (retic-count program). In addition, hematocrits are manually determined. The percent of corrected reticulocytes is determined using the following formula:
% RETICcORRECTED = % RETICoBSERVED X (HematθCπt,ND,viDUAL /
HematocπtwoRMAL) Active test compounds will show an increased % RETICCORRECTED level relative to mice that did not receive test peptides or EPO. Use of EPO-R agonist peptides of the invention
The peptide compounds of the invention are useful in vitro as tools for understanding the biological role of EPO, including the evaluation of the many factors thought to influence, and be influenced by, the production of EPO and the binding of EPO to the EPO-R (e g , the mechanism of EPO / EPO-R signal transduction/receptor activation). The present peptides are also useful in the development of other compounds that bind to the EPO-R, because the present compounds provide important structure-activity-relationship information that facilitate that development.
Moreover, based on their ability to bind to EPO-R, the peptides of the present invention can be used as reagents for detecting EPO-R on living cells; fixed cells; m biological fluids; in tissue homogenates; in purified, natural biological materials; etc. For example, by labeling such peptides, one can identify cells having EPO-R on their surfaces. In addition, based on their ability to bind EPO- R, the peptides of the present invention can be used in in situ staining, FACS (fluorescence-activated cell sorting) analysis, Western blotting, ELISA (enzyme-linked immunosorbent assay), etc. In addition, based on their ability to bind to EPO-R, the peptides of the present invention can be used m receptor purification, or in purifying cells expressing EPO-R on the cell surface (or inside permeabihzed cells)
The peptides of the invention can also be utilized as commercial reagents for various medical research and diagnostic purposes. Such uses can include but are not limited to: (1) use as a calibration standard for quantitating the activities of candidate EPO-R agonists in a variety of functional assays; (2) use as blocking reagents in random peptide screening, i.e , in looking for new families of EPO-R peptide hgands, the peptides can be used to block recovery of EPO peptides of the present invention; (3) use in co-crystalhzation with EPO-R, i e , crystals of the peptides of the present invention bound to the EPO-R may be formed, enabling determination of receptor/peptide structure by X-ray crystallography; (4) use to measure the capacity of erythrocyte precursor cells induce globm synthesis and heme complex synthesis, and to increase the number of ferritin receptors, by initiating differentiation; (5) use to maintain the proliferation and growth of EPO-dependent cell lines, such as the FDCP- 1-mEPO-R and the TF-I cell lines; and (6) other research and diagnostic applications wherein the EPO-R is preferably activated or such activation is conveniently calibrated against a known quantity of an EPO-R agonist, and the like. In yet another aspect of the present invention, methods of treatment and manufacture of a medicament are provided. The peptide compounds of the invention may be administered to warm blooded animals, including humans, to simulate the binding of EPO to the EPO-R in vivo. Thus, the present invention encompasses methods for therapeutic treatment of disorders associated with a deficiency of EPO, which methods comprise administering a peptide of the invention in amounts sufficient to stimulate the EPO-R and thus, alleviate the symptoms associated with a deficiency of EPO in vivo. For example, the peptides of this invention will find use in the treatment of renal insufficiency and/or end-stage renal failure/dialysis; anemia associated with AIDS; anemia associated with chronic inflammatory diseases (for example, rheumatoid arthritis and chronic bowel inflammation) and autoimmune disease; and for boosting the red blood count of a patient prior to surgery Other disease states, disorders, and states of hematologic irregularity that may be treated by administration of the peptides of this invention include, beta-thalassemia; cystic fibrosis; pregnancy and menstrual disorders, early anemia of prematurity; spinal cord injury, space flight; acute blood loss, aging, and various neoplastic disease states accompanied by abnormal erythropoiesis.
In other embodiments, the peptide compounds of the invention may be used for the treatment of disorders which are not characterized by low or deficient red blood cells, for example as a pretreatment prior to transfusions. In addition, administration of the compounds of this invention can result in a decrease in bleeding time and thus, will find use m the administration to patients prior to surgery or for indications wherein bleeding is expected to occur. In addition, the compounds of this invention will find use in the activation of megakaryoctes.
Since EPO has been shown to have a mitogenic and chemotactic effect on vascular endothelial cells as well as an effect on central cholinergic neurons [see, e g , Amagnostou, et al. (1990) Proc. Natl. Acad. Sci USA 87:5978-5982 and Konishi, et al. (1993) Brain Res. 609:29-35], the compounds of this invention will also find use for the treatment of a variety of vascular disorders, such as. promoting wound healing; promoting growth of collateral coronary blood vessels (such as those that may occur after myocardial infarction); trauma treatment; and post-vascular graft treatment. The compounds of this invention will also find use for the treatment of a variety of neurological disorders, generally characterized by low absolute levels of acetyl choline or low relative levels of acetyl choline as compared to other neuroactive substances e g., neurotransmitters.
Pharmaceutical compositions
In yet another aspect of the present invention, pharmaceutical compositions of the above EPO-R agonist peptide compounds are provided. Conditions alleviated or modulated by the administration of such compositions include those indicated above. Such pharmaceutical compositions may be for administration by oral, parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), transdermal (either passively or using iontophoresis or electroporation), transmucosal (nasal, vaginal, rectal, or sublingual) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate for each route of administration. In general, comprehended by the invention are pharmaceutical compositions comprising effective amounts of an EPO-R agonist peptide, or derivative products, of the invention together with pharmaceutically acceptable diluents, preservatives, solubihzers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents of various buffer content {eg , Tπs-HCl, acetate, phosphate), pH and ionic strength, additives such as detergents and solubilizing agents {e.g , Tween 20, Tween 80, Polysorbate 80), anti-oxidants (e g , ascorbic acid, sodium metabisulfite), preservatives (e g , Thimersol, benzyl alcohol) and bulking substances (e g , lactose, manmtol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc or into liposomes Hylauronic acid may also be used Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present proteins and derivatives. See, e g , Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712 which are herein incorporated by reference. The compositions may be prepared in liquid form, or may be in dried powder (e g , lyophihzed) form.
Oral Delivery
Contemplated for use herein are oral solid dosage forms, which are described generally in Remington's Pharmaceutical Sciences, 18th Ed. 1990 (Mack Publishing Co. Easton PA 18042) at Chapter 89, which is herein incorporated by reference. Solid dosage forms include tablets, capsules, pills, troches or lozenges, cachets, pellets, powders, or granules Also, liposomal or protemoid encapsulation may be used to formulate the present compositions (as, for example, protemoid microspheres reported in U S Patent No 4,925,673) Liposomal encapsulation may be used and the liposomes may be deπvatized with various polymers (e g , U S Patent No. 5,013,556) A descπption of possible solid dosage forms for the therapeutic is given by Marshall, K. In: Modern Pharmaceutics Edited by G S Banker and C T Rhodes Chapter 10, 1979, herein incorporated by reference In general, the formulation will include the EPO-R agonist peptides (or chemically modified forms thereof) and inert ingredients which allow for protection against the stomach environment, and release of the biologically active material in the intestine.
Also contemplated for use herein are liquid dosage forms for oral administration, including pharmaceutically acceptable emulsions, solutions, suspensions, and syrups, which may contain other components including inert diluents; adjuvants such as wetting agents, emulsifying and suspending agents; and sweetening, flavoring, and perfuming agents. The peptides may be chemically modified so that oral delivery of the derivative is efficacious.
Generally, the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of proteolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the component or components and increase in circulation time in the body. As discussed above, PEGylation is a preferred chemical modification for pharmaceutical usage Other moieties that may be used include: propylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrohdone, polyprohne, poly-l ,3-dioxolane and poly-l,3,6-tioxocane [see, e g , Abuchowski and Davis (1981) "Soluble Polymer-Enzyme Adducts," in Enzymes as Drugs Hocenberg and Roberts, eds (Wiley-Interscience: New York, NY) pp. 367-383; and Newmark, et al (1982) J Appl. Biochem. 4- 185-189]
For oral formulations, the location of release may be the stomach, the small intestine (the duodenum, the jejunem, or the ileum), or the large intestine One skilled in the art has available formulations which will not dissolve in the stomach, yet will release the material m the duodenum or elsewhere in the intestine. Preferably, the release will avoid the deleterious effects of the stomach environment, either by protection of the peptide (or derivative) or by release of the peptide (or derivative) beyond the stomach environment, such as in the intestine. To ensure full gastric resistance a coating impermeable to at least pH 5.0 is essential.
Examples of the more common inert ingredients that are used as enteric coatings are cellulose acetate tπmelhtate (CAT), hydroxypropylmethylcellulose phthalate (HPMCP), HPMCP 50, HPMCP 55, polyvinyl acetate phthalate (PVAP), Eudragit L30D, Aquateπc, cellulose acetate phthalate (CAP), Eudragit L, Eudragit S, and Shellac. These coatings may be used as mixed films. A coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach. This can include sugar coatings, or coatings which make the tablet easier to swallow Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic (ι e powder), for liquid forms a soft gelatin shell may be used. The shell mateπal of cachets could be thick starch or other edible paper. For pills, lozenges, molded tablets or tablet triturates, moist massing techniques can be used.
The peptide (or derivative) can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about lmm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs, or even as tablets. These therapeutics could be prepared by compression. Colorants and/or flavoring agents may also be included. For example, the peptide (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.
One may dilute or increase the volume of the peptide (or derivative) with an inert material These diluents could include carbohydrates, especially mannitol, α-lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride. Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell.
Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberhte, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used. The disintegrants may also be insoluble cationic exchange resins. Powdered gums may be used as disintegrants and as binders, and can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants Binders may be used to hold the peptide (or derivative) agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC) Polyvinyl pyrrolidone (PVP) and hydroxypropyl methyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the peptide (or derivative).
An antifrictional agent may be included in the formulation of the peptide (or derivative) to prevent sticking during the formulation process. Lubricants may be used as a layer between the peptide (or derivative) and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
Ghdants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The ghdants may include starch, talc, pyrogenic silica and hydrated silicoalummate To aid dissolution of the peptide (or derivative) into the aqueous environment a surfactant might be added as a wetting agent Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate. Cationic detergents might be used and could include benzalkomum chloride or benzethomium chloride. The list of potential nonionic detergents that could be included in the formulation as surfactants are lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 20, 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose These surfactants could be present in the formulation of the protein or derivative either alone or as a mixture in different ratios
Additives which potentially enhance uptake of the peptide (or derivative) are for instance the fatty acids oleic acid, linoleic acid and linolenic acid
Controlled release oral formulations may be desirable The peptide (or derivative) could be incorporated into an inert matrix which permits release by either diffusion or leaching mechanisms, e g , gums Slowly degenerating matrices may also be incorporated into the formulation. Some enteric coatings also have a delayed release effect Another form of a controlled release is by a method based on the Oros therapeutic system (Alza Corp.), i e the drug is enclosed in a semipermeable membrane which allows water to enter and push drug out through a single small opening due to osmotic effects.
Other coatings may be used for the formulation These include a variety of sugars which could be applied in a coating pan. The peptide (or derivative) could also be given in a film coated tablet and the materials used in this instance are divided into 2 groups The first are the nonenteπc materials and include methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl -methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols. The second group consists of the enteric materials that are commonly esters of phthahc acid.
A mix of materials might be used to provide the optimum film coating. Film coating may be carried out in a pan coater or in a fluidized bed or by compression coating.
Parenteral delivery
Preparations according to this invention for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use.
Rectal or vaginal delivery
Compositions for rectal or vaginal administration are preferably suppositories which may contain, in addition to the active substance, excipients such as cocoa butter or a suppository wax.
Compositions for nasal or sublingual administration are also prepared with standard excipients well known in the art.
Pulmonary Delivery
Also contemplated herein is pulmonary delivery of the EPO-R agonist peptides (or derivatives thereof). The peptide (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream [see, e g , Adjei, et al. (1990) Pharmaceutical Research 7:565-569; Adjei, et al. (1990) Int. J Pharmaceutics 63: 135-144 (leuprohde acetate); Braquet, et al. (1989) J. Cardiovascular Pharmacology 13(sup5): 143-146 (endothehn-1), Hubbard, et al (1989) Annals of Internal Medicine, Vol. Ill, pp. 206-212 (αl -antitrypsin); Smith, et al (1989) J. Clin. Invest 84.1 145-1 146 (α-1 -proteinase); Oswem, et al. (1990) "Aerosohzation of Proteins," Proceedings of Symposium on Respiratory Drug Delivery II Keystone, Colorado (recombinant human growth hormone); Debs, et al. (1988) J. Immunol. 140:3482-3488 (interferon-γ and tumor necrosis factor α), and U.S. Pat. No. 5,284,656 to Platz, et al. (granulocyte colony stimulating factor). A method and composition for pulmonary delivery of drugs for systemic effect is described in U S Pat. No. 5,451 ,569 to Wong, et al. Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer (Mallinckrodt Inc , St. Louis, MO); the Acorn II nebulizer (Marquest Medical Products, Englewood, CO), the Ventolin metered dose inhaler (Glaxo Inc , Research Triangle Park, NC), and the Spmhaler powder inhaler (Fisons Corp , Bedford, MA).
All such devices require the use of formulations suitable for the dispensing of peptide (or derivative) Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated Chemically modified peptides may also be prepared in different formulations depending on the type of chemical modification or the type of device employed
Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise peptide (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active protein per mL of solution The formulation may also include a buffer and a simple sugar (e g , for protein stabilization and regulation of osmotic pressure). The nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the peptide (or derivative) caused by atomization of the solution in forming the aerosol
Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the peptide (or derivative) suspended in a propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including tπchlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and
1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin Oleic acid may also be useful as a surfactant.
Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing peptide (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e g , 50 to 90% by weight of the formulation. The peptide (or derivative) should most advantageously be prepared in particulate form with an average particle size of less than 10 mm (or microns), most preferably 0.5 to 5 mm, for most effective delivery to the distal lung.
Nasal Delivery
Nasal delivery of the EPO-R agonist peptides (or derivatives) is also contemplated. Nasal delivery allows the passage of the peptide to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung Formulations for nasal delivery include those with dextran or cyclodextran. Other penetration-enhancers used to facilitate nasal delivery are also contemplated for use with the peptides of the present invention (such as described in International Patent Publication No. WO 2004056314, filed December 17, 2003, incorporated herein by reference in its entirety).
Dosages
For all of the peptide compounds, as further studies are conducted, information will emerge regarding appropriate dosage levels for treatment of various conditions in various patients, and the ordinary skilled worker, considering the therapeutic context, age, and general health of the recipient, will be able to ascertain proper dosmg The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. Generally dosage levels of 0.001 to 10 mg/kg of body weight daily are administered to mammals. Generally, for intravenous injection or infusion dosage may be lower. The dosing schedule may vary, depending on the circulation half-life, and the formulation used.
The peptides of the present invention (or their derivatives) may be administered in conjunction with one or more additional active ingredients or pharmaceutical compositions.
Examples
The present invention is next described by means of the following examples. However, the use of these and other examples anywhere in the specification is illustrative only, and in no way limits the scope and meaning of the invention or of any exemplified form. Likewise, the invention is not limited to any particular preferred embodiments described herein. Indeed, many modifications and variations of the invention may be apparent to those skilled in the art upon reading this specification, and can be made without departing from its spirit and scope. The invention is therefore to be limited only by the terms of the appended claims, along with the full scope of equivalents to which the claims are entitled.
The listed examples describe experiments by which someone of ordinary skill m the art may ascertain the biological activity of the peptides of present invention.
Example 1: Synthesis of EPO-R agonist peptides This example describes preferred, non-limiting embodiments of methods by which peptides covered by the present invention can be synthesiaed. However, other methods, which have been previously described for the synthesis of EPO peptide moieties (see, for example, in PCT/US04/14886, filed May 12, 2004) can also be used to prepare compounds of this invention
Solid phase techniques are provided for synthesizing both peptide monomers and dimers of the invention. Exemplary techniques for attaching linker and PEG moieties to a peptide compound of this invention are also described, as well as methods for oxidizing the peptide compounds, e.g., forming intramolecular disulfide bonds. Finally, this example also provides a technique for purifying peptide compounds that are synthesized according to these methods.
1 Peptide Monomer Synthesis Various peptide monomers of the invention can be synthesized, as described here, using the
Merπfield solid phase synthesis technique [see, Stewart and Young. Solid Phase Peptide Synthesis, 2nd edition (Pierce Chemical, Rockford, IL) 1984] on an Applied Biosystems 433 A automated instrument. The resin PAL (Milhgen/Biosearch) is used, which is cross-linked polystyrene with 5-(4'- Fmoc-aminomethyl-3,5'-dimethoxyphenoxy) valeric acid. Use of PAL resm results in a carboxyl terminal amide functional group upon cleavage of the peptide from the resin Primary amine protection on amino acids is achieved with Fmoc, and side chain protection groups is t-butyl for serine, threonine, and tyrosine hydroxyls; tπtyl for glutamine and asparagme amides; Trt or Acm for cysteine; and PMC (2,2,5,7,8-pentamethylchroman sulfonate) for the argimne guanidmo group. Each coupling is performed for either 1 hr or 2 hr with BOP (benzotriazolyl N- oxtπsdimethylammophosphonium hexafluorophosphate) and HOBt (1 -hydroxybenztriazole).
For the synthesis of peptides with an amidated carboxy terminus, the fully assembled peptide is cleaved with a mixture of 90% trifluoroacetic acid, 5% ethanedithiol, and 5% water, initially at 4°C and gradually increased to room temperature over 1.5 hr. The deprotected product is filtered from the resm and precipitated with diethyl ether. After thorough drying the product is purified by Cl 8 reverse phase high performance liquid chromatography with a gradient of acetonitπle/water in 0 1% trifluoroacetic acid.
2 Peptide Dimer Synthesis
Various peptide dimers of the invention are synthesized directly onto a lysine linker in a variation of the solid phase technique.
For simultaneous synthesis of the two peptide chains, Fmoc-Lys(Fmoc)-OH is coupled to a PAL resin (Milhgen/Biosearch), thereby providing an initial lysine residue to serve as the linker between the two chains to be synthesized The Fmoc protecting groups are removed with mild base (20% pipeπdme in DMF), and the peptide chains are synthesized using the resulting free ammo groups as starting points. Peptide chain synthesis is performed using the solid phase synthesis technique described above. Trt is used to protect all cysteine residues. Following dimer deprotection, cleavage from the resin, and purification, oxidation of the cysteine residues is performed by incubating the deprotected dimer in 100% DMSO for 2-3 days at 5°C to 25°C. This oxidation reaction can yield predominantly (>75%) dimers with two intramolecular disulfide bonds. For sequential synthesis of the two peptide chains, Fmoc-Lys(AUoc)-OH is coupled to a PAL resin (Milhgen/Biosearch), thereby providing an initial lysine residue to serve as the linker between the two chains to be synthesized. The Fmoc protecting group is removed with mild base (20% pipeπdine in DMF) The first peptide chain is then syntheszed using the resulting free amino group as a starting point Peptide synthesis is performed using the solid phase technique described above The two cysteine residues of the first chain are protected with Trt Following synthesis of the first peptide chain, the Alloc group is removed from the support-bound lysine linker with Pd[P(C6H5)3]4, 4-methyl morphohne, and chloroform The second peptide chain is then synthesized on this second free amino group The two cysteine residues of the second chain are protected with Acm An intramolecular disulfide bond is formed in the first peptide chain by removing the Trt protecting groups using tπfluoroacetic acid, followed by oxidation by stirring in 20% DMSO overnight An intramolecular disulfide bond is then formed m the second peptide chain by simultaneously removing the Acm protecting groups and oxidizing the deprotected cysteine residues using iodine, methanol, and thahum tπfluoroacetate
3 Attachment of spacers
Where the spacer is an ammo acid (e g , glycine or lysine), the spacer is incorporated into the peptide during solid phase peptide synthesis In this case, the spacer amino acid is coupled to the PAL resin, and its free ammo group can serve as the basis for the attachment of another spacer ammo acid, or of the lysine linker Following the attachment of the lysine linker, dimeric peptides are synthesized as described above
4 Oxidation of peptides to form intramolecular disulfide bonds
The peptide dimer is dissolved in 20% DMSO/water (1 mg dry weight peptide/mL) and is allowed to stand at room temperature for 36h The peptide is purified by loading the reaction mixture onto a Cl 8 HPLC column (Waters Delta-Pak Cl 8, 15 micron particle size, 300 angstrom pore size, 40 mm x 200 mm length), followed by a linear ACN/water/0 01% TFA gradiant from 5 to 95% ACN over 40 minutes Lyophohzation of the fractions containing the desired peptide affords the product as a fluffy white solid
5 PEGylatwn of peptides PEGylation of the peptides of the invention can be carried out using several different techniques
PEGylation of a terminal -NH2 group: The peptide dimer is mixed with 1 5 eq (mole basis) of activated PEG species (mPEG-NPC from NOF Corp Japan) in dry DMF to afford a clear solution
After 5 minutes 4eq of DIEA is added to the above solution The mixture is stirred at ambient temperature 14h, followed by purification with C18 reverse phase HPLC The structure of PEGylated peptide is confirmed by MALDI mass The purified peptide is also subjected to purification via cation ion exchange chromatography as outlined below DiPEGylation of the N-termini of a peptide dimer: The peptide dimer is mixed with 2.5 eq.
(mole basis) of activated PEG species (mPEG-NPC from NOF Corp. Japan) in dry DMF to afford a clear solution. After 5 minutes 4eq of DIEA is added to the above solution. The mixture is stirred at ambient temperature 14h, followed by purification with Cl 8 reverse phase HPLC. The purified peptide is also subjected to purification via cation ion exchange chromatography as outlined below.
Peptide dimerization via PEGylation of N-termini: The peptide (2.5 eq.) and PEG-(SPA-
NHS)2 (1 eq from Shearwater Corp, USA.) is dissolved in dry DMF at 0.25M to afford a clear solution After 5 minutes lOeq of DIEA is added to the above solution. The mixture is stirred at ambient temperature 2h, followed by purification with Cl 8 reverse phase HPLC. The purified peptide is also subjected to purification via cation ion exchange chromatography as outlined below.
Peptide dimerization via PEGylation of C-termini: The peptide (2.5 eq.) and PEG-(SPA-
NHS)2 (1 eq. from Shearwater Corp, USA.) is dissolved in dry DMF at 0.25M to afford a clear solution. After 5 minutes lOeq of DIEA is added to the above solution. The mixture is stirred at ambient temperature 2h, followed by purification with C18 reverse phase HPLC. The purified peptide is also subjected to purification via cation ion exchange chromatography as outlined below.
6 Ion exchange purification of peptides
Several exchange supports can be surveyed for their ability to separate the above peptide-PEG conjugate from unreacted (or hydrolyzed) PEG, in addition to their ability to retain the starting dimeπc peptides. The ion exchange resm (2-3g) is loaded into a 1 cm column, followed by conversion to the sodium form (0.2 N NaOH loaded onto column until elutant was pH 14, ca. 5 column volumes), and then to the hydrogen form (eluted with either 0.1 N HCl or 0.1 M HOAc until elutant matched load pH, ca. 5 column volumes), followed by washing with 25% ACN/water until pH 6. Either the peptide prior to conjugation or the peptide-PEG conjugate is dissolved in 25% ACN/water (10 mg/mL) and the pH adjusted to <3 with TFA, then loaded on the column. After washing with 2-3 column volumes of 25% ACN/water and collecting 5 mL fractions, the peptide is released from the column by elution with 0 1 M NH4OAc in 25% ACN/water, again collecting 5 mL fractions. Analysis via HPLC can reveal which fractions contain the desired peptide. Analysis with an Evaporative Light- Scattering Detector (ELSD) can indicate that when the peptide is retained on the column and is eluted with the NH4OAc solution (generally between fractions 4 and 10), no non-conjugated PEG is observed as a contaminant. When the peptide elutes in the initial wash buffer (generally the first 2 fractions), no separation of desired PEG-conjugate and excess PEG may be observed.
The following columns can possibly successfully retain both the peptide and the peptide-PEG conjugate, and successfully purify the peptide-PEG conjugate from the unconjugates peptide:
Ion Exhange Resins
Figure imgf000046_0001
Figure imgf000047_0001
Example 2: In vitro activity assays
This example describes certain in vitro assays that are useful for evaluating the activity and potency of peptides covered by this invention, e.g., as EPO-R agonists. In particular, the results obtained from assays such as the ones described here demonstrate whether a peptide compound binds to EPO-R and activates EPO-R signalling. The assays can also be used to compare the binding efficiency and biological activity of a compound, for example, to other, known EPO mimetic compounds
EPO-R agonist peptide monomers and dimers tested in these assays are typically prepared according to methods such as those described in Example 1. The potency of these peptide monomers and dimers is then evaluated using a series of in vitro activity assays, including: a reporter assay, a proliferation assay, a competitive binding assay, and a C/BFU-e assay. These four assays are described in further detail below.
1. Reporter assay
This assay is based upon a on a murine pre-B-cell line derived reporter cell, Baf3/EpoR/GCSFR fos/lux. This reporter cell line expresses a chimeric receptor comprising the extracellular portion of the human EPO receptor to the lntra-cellular portion of the human GCSF receptor. This cell line is further transfected with a fos promoter-driven luciferase reporter gene construct. Activation of this chimeric receptor through addition of erythropoietic agent results in the expression of the luciferase reporter gene, and therefore the production of light upon addition of the luciferase substrate lucifeπn. Thus, the level of EPO-R activation in such cells may be quantitated via measurement of luciferase activity.
The Baf3/EpoR/GCSFR fos/lux cells are cultured in DMEM/F12 medium (Gibco) supplemented with 10% fetal bovine serum (FBS; Hyclone), 10% WEHI-3 supernatant (the supernatant from a culture of WEHI-3 cells, ATCC # TIB-68), and penicillin/streptomycin. Appioximately 18 h before the assay, cells are starved by transferring them to DMEM/F12 medium supplemented with 10% FBS and 0.1% WEHI-3 supernatant. On the day of assay, cells are washed once with DMEM/F12 medium supplemented with 10% FBS (no WEHI-3 supernatant), then 1 X 106 cells/mL are cultured in the presence of a known concentration of test peptide, or with EPO (R & D Systems Inc., Minneapolis, MN) as a positive control, in DMEM/F12 medium supplemented with 10% FBS (no WEHI-3 supernatant). Serial dilutions of the test peptide are concurrently tested in this assay Assay plates are incubated for 4h at 370C in a 5% CO2 atmosphere, after which lucifeπn (Steady-Glo; Promega, Madison, WI) is added to each well Following a 5-minute incubation, light emission is measured on a Packard Topcount Luminometer (Packard Instrument Co., Downers Grove, 111.)- Light counts are plotted relative to test peptide concentration and analysed using Graph Pad software. The concentration of test peptide that results in a half-maximal emission of light is recorded as the EC50.
2 Proliferation assay
This assay is based upon a murine pre-B-cell line, BaO, transfected to express human EPO-R. Proliferation of the resulting cell line, BaF3/Gal4/Elk/EPOR, is dependent on EPO-R activation. The degree of cell proliferation is quantitated using MTT, where the signal in the MTT assay is proportional to the number of viable cells. The BaF3/Gal4/Elk/EPOR cells are cultured in spinner flasks m DMEM/F12 medium (Gibco) supplemented with 10% FBS (Hyclone) and 2% WEHI-3 supernatant (ATCC # TBB-68). Cultured cells are starved overnight, in a spmner flask at a cell density of IxIO6 cells/ml, in DMEM/F12 medium supplemented with 10% FBS and 0.1% WEHI-3 supernatant. The starved cells are then washed twice with Dulbecco's PBS (Gibco), and resuspended to a density of 1x106 cells/ml in DMEM/F12 supplemented with 10% FBS (no WEHI-3 supernatant). 50μL ahquots (-50,000 cells) of the cell suspension are then plated, in triplicate, in 96 well assay plates. 50μL ahquots of dilution series of test EPO mimetic peptides, or 50μL EPO (R & D Systems Inc., Minneapolis, MN) or Aranesp™ (darbepoeitm alpha, an ERO-R agonist commeπcally available from Amgen) in DMEM/F12 media supplemented with 10% FBS (no WEHI-3 supernatant I) are added to the 96 well assay plates (final well volume of lOOμL). For example, 12 different dilutions may be tested where the final concentration of test peptide (or control EPO peptide) ranges from 81OpM to 0.0045pM. The plated cells are then incubated for 48h at 370C. Next, lOμL of MTT (Roche Diagnostics) is added to each culture dish well, and then allowed to incubate for 4h. The reaction is then stopped by adding 10% SDS + 0.01N HCl The plates are then incubated overnight at 370C. Absorbance of each well at a wavelenght of 595nm is then measured by spectrophotometry. Plots of the absorbance readings versus test peptide concentration are constructed and the EC50 calculated using Graph Pad software. The concentration of test peptide that results in a half-maximal absorbance is recorded as the EC50.
3. Competitive Binding Assay Competitive binding calculations are made using an assay in which a light signal is generated as a function of the proximity of two beads: a streptavidin donor bead bearing a biotinylated EPO-R- binding peptide tracer and an acceptor bead to which is bound EPO-R. Light is generated by non- radiative energy transfer, during which a singlet oxygen is released from a first bead upon illumination, and contact with the released singlet oxygen causes the second bead to emit light. These bead sets are commercially available (Packard). Bead proximity is generated by the binding of the EPO-R-binding peptide tracer to the EPO-R A test peptide that competes with the EPO-R-binding peptide tracer for binding to EPO-R will prevent this binding, causing a decrease in light emission
In more detail the method is as follows: Add 4μL of serial dilutions of the test EPO-R agonist peptide, or positive or negative controls, to wells of a 384 well plate. Thereafter, add 2μL / well of receptor/bead cocktail. Receptor bead cocktail consists of 15μL of 5mg/ml streptavidin donor beads (Packard), 15μL of 5mg/ml monoclonal antibody abl79 (this antibody recognizes the portion of the human placental alkaline phosphatase protein contained in the recombinant EPO-R), protein A-coated acceptor beads (protein A will bind to the abl79 antibody; Packard), 112.5μL of a 1 :6.6 dilution of recombinant EPO-R (produced in Chinese Hamster Ovary cells as a fusion protein to a portion of the human placental alkaline phosphatase protein which contains the abl79 target epitope) and 607.5 μL of Alphaquest buffer (4OmM HEPES, pH 7.4; ImM MgCl2; 0.1% BSA, 0.05% Tween 20). Tap to mix. Add 2μL/well of a biotinylated EPO-R-bmding peptide tracer.
Centrifuge 1 min to mix. Seal plate with Packard Top Seal and wrap m foil Incubate overnight at room temperature. After 18 hours read light emission using an AlphaQuest reader (Packard) Plot light emission vs concentration of peptide and analyse with Graph Pad or Excel.
The concentration of test peptide that results in a 50% decrease in light emission, relative to that observed without test peptide, is recorded as the IC50.
4 C/BFU-e Assay EPO-R signaling stimulates the differentiation of bone marrow stem cells into proliferating red blood cell presursors. This assay measures the ability of test peptides to stimulate the proliferation and differentiation of red blood cell precursors from primary human bone marrow pluπpotent stem cells.
For this assay, serial dilutions of test peptide are made in EVIDM medium (Gibco) supplemented with 10% FBS (Hyclone). These serial dilutions, or positive control EPO peptide, are then added to methylcellulose to give a final volume of 1.5mL. The methylcellulose and peptide mixture is then vortexed thoroughly. Ahquots (100,000 cells/mL) of human, bone marrow deπved CD34+ cells (Poietics/Cambrex) are thawed. The thawed cells are gently added to 0 1 mL of lmg/ml DNAse (Stem Cells) in a 5OmL tube. Next, 40-50 mL IMDM medium is added gently to cells: the medium is added drop by drop along the side of the 5OmL tube for the first 1OmL, and then the remaining volume of medium is slowly dispensed along the side of the tube The cells are then spun at 900rpm for 20 mm, and the media removed carefully by gentle aspiration. The cells are resuspended in ImI of IMDM medium and the cell density per mL is counted on hemacytometer slide (lOμL aliquot of cell suspension on slide, and cell density is the average count X 10,000 cells/ml). The cells are then diluted in IMDM medium to a cell density of 15,000 cells/mL. A lOOμL of diluted cells is then added to each 1.5 mL methyl cellulose plus peptide sample (final cell concentration in assay media is 1000 cells/ mL), and the mixture is vortexed. Allow the bubbles in the mixture to disappear, and then aspirate ImL using blunt-end needle. Add 0.25 mL aspirated mixture from each sample into each of 4 wells of a 24-well plate (Falcon brand). Incubate the plated mixtures at 370C under 5% CO2 in a humid incubator for 14 days. Score for the presence of erythroid colonies using a phase microscope (5X- 1OX objective, final magnification of 100X). The concentration of test peptide at which the numer of formed colonies is 90% of maximum, relative to that observed with the EPO positive control, is recorded as the EC90. 5. Radioligand Competitive Binding Assay
An alternative radioligand competition binding assay can also be used to measure IC50 values for peptides of the present invention.. This assay measures binding of 125I-EPO to EPOr. The assay may be performed according to the following exemplary protocol: A. Materials
Figure imgf000050_0001
B. Determination of appropriate receptor concentration.
One 50 μg vial of lyophihzed recombinant EPOR extracellular domain fused to the Fc portion of human IgGl is reconstituted in 1 mL of assay buffer. To determine the correct amount of receptor to use in the assay, 100 μL serial dilutions of this receptor preparation are combined with approximately 20,000 cpm in 200 μL of iodinated recombinant human Erythropoietin (125I-EPO) in 12 x 75 mm polypropylene test tubes Tubes are capped and mixed gently at 4 0C overnight on a LabQuake rotating shaker
The next day, 50 μL of a 50 % slurry of Protein-G Sepharose is added to each tube Tubes are then incubated for 2 hours at 4 0C, mixing gently The tubes are then centrifuged for 15 min at 4000 RPM (3297 x G) to pellet the protein-G sepharose The supernatants are carefully removed and discarded After washing 3 times with 1 mL of 4 0C assay buffer, the pellets are counted in a Wallac Wizard gamma counter Results were then analyzed and the dilution required to reach 50 % of the maximum binding value was calculated
C IC50 Determination for peptide
To determine the IC50 of a peptide of the present invention, 100 μL serial dilutions of the peptide are combined with 100 μL of recombinant erythropoietin receptor (100 pg/tube) in 12 x 75 mm polypropylene test tubes Then 100 μL of iodmated recombinant human Erythropoietin (125I- EPO) is added to each tube and the tubes were capped and mixed gently at 40C overnight The next day, bound '25I-EPO is quantitated as described above The results are analyzed and the IC50 value calculated using Graphpad Prism version 4 0, from GraphPad Software, Lie (San Diego, CA) The assay is preferably repeated 2 or more times for each peptide whose IC50 value is measured by this procedure, for a total of 3 replicate IC50 determinations
Example 3: In vivo activity assays
This example describes certain in vivo assays that are useful for evaluating the activity and potency of peptides covered by this invention, e g , as EPO-R agonists In particular, the results obtained from assays such as the ones described here demonstrate whether a peptide compound binds to EPO-R and activates EPO-R signalling The assays can also be used to compare the binding efficiency and biological activity of a compound, for example, to other, known EPO mimetic compounds
This example describes various in vivo assays that are useful in evaluating the activity and potency of EPO-R agonist peptides of the invention EPO-R agonist peptide monomers and dimers tested in these assays are typically prepared according to the methods described in Example 1 The in vivo activity of these peptide monomers and dimers is then evaluated using a series assays, including a polycythemic exhypoxic mouse bioassay and a reticulocyte assay These two assays are described in further detail below
1 Polycythemic Exhypoxic Mouse Bioassay Test peptides are assayed for in vivo activity in the polycythemic exhypoxic mouse bioassay adapted from the method described by Cotes and Bangham (1961), Nature 191 ' 1065-1067. This assay examines the ability of a test peptide to function as an EPO mimetic: i e , to activate EPO-R and induce new red blood cell synthesis Red blood cell synthesis is quantitated based upon incorporation of radiolabeled iron into hemoglobin of the synthesized red blood cells.
BDFl mice are allowed to acclimate to ambient conditions for 7-10 days. Body weights are determined for all animals, and low weight animals (<15 grams) are not used. Mice are subjected to successive conditioning cycles in a hypobaπc chamber for a total of 14 days. Each 24 hour cycle consista of 18 hr at 0.40±0.02% atmospheric pressure and 6 hr at ambient pressure After conditioning the mice are maintained at ambient pressure for an additional 72 hr prior to dosing.
Test peptides, or recombinant human EPO standards, are diluted in PBS + 0.1% BSA vehicle
(PBS/BSA). Peptide monomer stock solutions are first solubihzed in dimethyl sulfoxide (DMSO).
Negative control groups include one group of mice injected with PBS/BSA alone, and one group injected with 1% DMSO Each dose group containa 10 mice. Mice are injected subcutaneously (scruff of neck) with 0 5 mL of the appropriate sample.
Forty eight hours following sample injection, the mice are administered an intraperitoneal injection of 0.2 ml of Fe59 (Dupont, NEN), for a dose of approximately 0.75 μCuπes/mouse. Mouse body weights are determined 24hr after Fe59 administration, and the mice are sacrificed 48hr after Fe59 administration. Blood is collected from each animal by cardiac puncture and hematocrits are determined (heparin was used as the anticoagulant). Each blood sample (0.2 ml) is analyzed for Fe59 incorporation using a Packard gamma counter. Non-responder mice (ι e , those mice with radioactive incorporation less than the negative control group) are eliminated from the appropriate data set. Mice that have hematocrit values less than 53% of the negative control group are also eliminated
Results are derived from sets of 10 animals for each experimental dose. The average amount of radioactivity incorporated [counts per mmute (CPM)] into blood samples from each group is calculated.
2. Reticulocyte Assay
Normal BDFl mice are dosed (0.5 mL, injected subcutaneously) on three consecutive days with either EPO control or test peptide. At day three, mice are also dosed (0.1 mL, injected lntrapeπtoneally) with iron dextran (100 mg/ml). At day five, mice are anesthetized with CO2 and bled by cardiac puncture. The percent (%) reticulocytes for each blood sample is determined by thiazole orange staining and flow cytometer analysis (retic-count program). Hematocrits are manually determined. The corrected percent of reticulocytes is determined using the following formula: % RETICcoRRECTED = % RETICoBSERVED X (HematocπtIND,viDUAL / HematocπtNORMAL)
3. Hematological Assay Normal CDl mice are dosed with four weekly bolus intravenous injections of either EPO positive control, test peptide, or vehicle. A range of positive control and test peptide doses, expressed as mg/kg, are tested by varying the active compound concentration in the formulation. Volumes injected are 5ml/kg The vehicle control group is comprised twelve animals, while 8 animals are in each of the remaining dose groups. Daily viability and weekly body weights are recorded.
The dosed mice are mice are fasted and then anesthetized with inhaled isoflurane and terminal blood samples are collected via cardiac or abdominal aorta puncture on Day 1 (for vehicle control mice) and on Days 15 and 29 (4 mice/group/day). The blood is transferred to Vacutainer® brand tubes. Preferred anticoagulant is ethylenediaminetetraacetic acid (EDTA). Blood samples are evaluated for endpoints measuring red blood synthesis and physiology such as hematocrit (Hct), hemoglobin (Hgb) and total erythrocyte count (RBC) using automated clinical analysers well known in the art (e g , those made by Coulter, Inc.).
The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figure(s). Such modifications are intended to fall within the scope of the appended claims. It is further to be understood that all values are approximate, and are provided for description.
Numerous references, including patents, patent applications, and various publications are cited and discussed throught the specification. The citation and/or discussion of such references is provided merely to clarify the description of the present invention and is not an admission that any such reference is "prior art" to the present invention All references cited and discussed in this specification are incorporated herein by reference in their entirety and to the same extent as if each reference was individually incorporated by reference.

Claims

WHAT IS CLAIMED IS:
1. A peptide, comprising an ammo acid sequence selected from SEQ ID NOS: 1-726 according to Figures IA - IL.
2. A peptide according to claim 1, wherein the N-terminal of said peptide is acetylated.
3. A peptide according to claim 1, wherein the peptide is a monomer.
4. A peptide according to claim 1, wherein the peptide is a dimer.
5. A peptide according to claim 4, wherein the peptide is a homodimer.
6. A peptide according to claim 1, further comprising one or more water soluble polymers covalently bound to the peptide.
7. A peptide according to claim 6, wherein the water soluble polymer is polyethylene glycol (PEG).
8 A peptide according to claim 7, wherein said PEG comprises a linear unbranched molecule having a molecular weight of about 500 to about 60,000 Daltons.
9. A peptide according to claim 8, wherein the PEG has a molecular weight of less than about 20,000 Daltons.
10. A peptide according to claim 8, wherein the PEG has a molecular weight of about 20,000 to about 60,000 Daltons.
1 1. A peptide according to claim 8, wherein the PEG has a molecular weight of about 20,000 to about 40,000 Daltons
12. A peptide according to claim 8, wherein two PEG moieities are covalently bound to the peptide, each of said PEG comprising a linear unbranched molecule
13 A peptide according to claim 12, wherein each of said PEG has a molecular weight of about 20,000 to about 30,000 Daltons
14. A peptide of claim 1, wherein said peptide binds to and activates the erythropoietin receptor (EPO-R).
15. A peptide dimer, comprising-
(a) a first peptide chain;
(b) a second peptide chain; and
(c) a linking moiety connecting said first and second peptide chains, wherein at least one of said first peptide chain and said second peptide chain comprises an amino acid sequence selected from SEQ ID NOS: 1-726 according to Figures IA - IL; and wherein said peptide binds to and activates the erythropoietin receptor (EPO-R).
16 A peptide dimer according to claim 15, wherein the linking moiety comprises the formula:
-NH-R3-NH- wherein R3 is a lower (C|.6) alkylene.
17 A peptide dimer according to claim 16, wherein the linking moiety is a lysine residue.
18. A peptide dimer according to claim 15, wherein the linking moiety comprises the formula:
-CO-(CH2),,-X-(CH2)m-CO- wherein n is an integer from 0 to 10, m is an integer from 1 to 10, X is selected from O, S, N(CH2)pNR,, NCO(CH2)pNRi, and CHNRi , R1 is selected from H, Boc, and Cbz, and p is an integer from l to 10
19. A peptide dimer according to claim 18, wherein n and m are each 1, X is NCO(CH2)PNR1, p is 2, and R1 is H.
20. A peptide dimer according to claim 15, further comprising a water soluble polymer.
21. A peptide dimer according to claim 20, wherein the water soluble polymer is covalently bound to the linker moiety.
22. A peptide dimer according to claim 15, further comprising a spacer moiety.
23. A peptide dimer according to claim 22, wherein the spacer moiety comprises the formula:
-NH-(CH2)α-[O-(CH2)p]γ-Oδ-(CH2)6-Y- wherein α, β, and ε are each integers whose values are independently selected from 1 to 6, δ is 0 or 1, γ is an integer selected from 0 to 10, and Y is selected from NH or CO, provided that β is 2 when γ is greater than 1
24. A peptide dimer according to claim 23 wherein each of α, β, and ε is 2, each of γ and δ is 1, and Y is NH.
25. A peptide dimer according to claim 22, further comprising one or more water soluble polymers.
26. A peptide dimer according to claim 25, wherein the water soluble polymer is covalently bound to the spacer moiety.
27 A peptide dimer according to claim 20 or 25, wherein the water soluble polymer is polyethylene glycol (PEG).
28. A peptide dimer according to claim 27, wherein the PEG is a linear unbranched PEG having a molecular weight of about 500 to about 60,000 Daltons.
29. A peptide dimer according to claim 28, wherein the PEG has a molecular weight of about 500 to less than about 20,000 Daltons.
30 A peptide dimer according to claim 28, wherein the PEG has a molecular weight of about 20,000 to 60,000 Daltons.
31. A peptide dimer according to claim 30, wherein the PEG has a molecular weight of about 20,000 to about 40,000 Daltons.
32. A peptide according to claim 27, wherein two PEG moieities are covalently bound to the peptide, each of said PEG comprising a linear unbranched molecule.
33. A peptide according to claim 32, wherein each of said PEG has a molecular weight of about 20,000 to about 30,000 Daltons.
34 A method for treating a patient, comprising administering to a patient having a disorder characterized by a deficiency of erythropoietin or a low or defective red blood cell population a therapeutically effective amount of a peptide comprising an amino acid sequence selected from SEQ ID NOS: 1-726 according to Figures IA - IL.
35. A method according to claim 34, wherein the disorder is selected from: end stage renal failure or dialysis; anemia associated with AIDS, auto immune disease or a malignancy; beta-thalassemia; cystic fibrosis; early anemia of prematurity; anemia associated with chronic inflammatory disease; spinal cord injury; acute blood loss; aging; and neoplastic disease states accompanied by abnormal erythropoiesis.
36 A method according to claim 34, wherein the peptide is a monomer.
37. A method according to claim 34, wherein the peptide is a dimer.
38. A method according to claim 37, wherein the peptide is a homodimer.
39. A method according to claim 34, wherein one or more water soluble polymers are covalently bound to the peptide.
40. A method according to claim 39, wherein the water soluble polymer is polyethylene glycol (PEG)
41. A method according to claim 40, wherein the PEG is a linear unbranched PEG having a molecular weight of about 500 to about 60,000 Daltons.
42. A method according to claim 41, wherein the PEG has a molecular weight of about 500 to less than about 20,000 Daltons.
43 A peptide dimer according to claim 41, wherein the PEG has a molecular weight of about 20,000 to 60,000 Daltons.
44. A method according to claim 43, wherein the PEG has a molecular weight of about 20,000 to about 40,000 Daltons.
45. A method according to claim 40, wherein two PEG moieities are covalently bound to the peptide, each of said PEG comprising a linear unbranched molecule.
46. A method according to claim 45, wherein each of said PEG has a molecular weight of about 20,000 to about 30,000 Daltons
47. A pharmaceutical composition comprising:
(i) a peptide comprising an ammo acid sequence selected from SEQ ID NOS: 1-726 according to Figures IA - IL; and (ii) a pharmaceutically acceptable carrier.
48. A pharmaceutical composition according to claim 47, wherein the peptide is a monomer.
49. A pharmaceutical composition according to claim 47, wherein the peptide is a dimer.
50 A pharmaceutical composition according to claim 49, wherein the peptide is a homodimer.
51. A pharmaceutical composition according to claim 50, wherein one or more water soluble polymers is covalently bound to the peptide.
52. A pharmaceutical composition according to claim 51 , wherein the water soluble polymer is polyethylene glycol (PEG).
53 A pharmaceutical composition according to claim 52, wherein the PEG is a linear unbranched PEG having a molecular weight of about 500 to about 60,000 Dal tons.
54 A pharmaceutical composition according to claim 53, wherein the PEG has a molecular weight of about 500 to less than about 20,000 Daltons.
55. A pharmaceutical composition according to claim 53, wherein the PEG has a molecular weight of about 20,000 to about 60,000 Daltons.
56. A pharmaceutical composition according to claim 55, wherein the PEG has a molecular weight of about 20,000 to about 40,000 Daltons.
57. A pharmaceutical composition according to claim 52, wherein two PEG moieties are covalently bound to the peptide, each of said PEG comprising a linear unbranched molecule.
58. A pharmaceutical composition according to claim 57, wherein each of said PEG has a molecular weight of about 20,000 to 30,000 Daltons.
PCT/US2005/041112 2004-11-11 2005-11-11 Novel peptides that bind to the erythropoietin receptor WO2006062685A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62743204P 2004-11-11 2004-11-11
US60/627,432 2004-11-11

Publications (2)

Publication Number Publication Date
WO2006062685A2 true WO2006062685A2 (en) 2006-06-15
WO2006062685A3 WO2006062685A3 (en) 2006-10-19

Family

ID=36578375

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/041112 WO2006062685A2 (en) 2004-11-11 2005-11-11 Novel peptides that bind to the erythropoietin receptor

Country Status (2)

Country Link
US (3) US20080108564A1 (en)
WO (1) WO2006062685A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008104513A1 (en) * 2007-03-01 2008-09-04 Novo Nordisk A/S Expression of proteins in e. coli
WO2009079910A1 (en) * 2007-12-12 2009-07-02 Jiangsu Hansen Pharmaceutical Co., Ltd. An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
EP2118127A2 (en) * 2007-01-31 2009-11-18 Affymax, Inc. Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
US7858747B2 (en) 2005-08-30 2010-12-28 Novo Nordisk A/S Expression of proteins in E.coli
US20110281791A1 (en) * 2009-01-28 2011-11-17 Zion Todd C Crystalline insulin-conjugates
US20110301083A1 (en) * 2009-01-28 2011-12-08 Zion Todd C Conjugate based systems for controlled drug delivery
WO2012054582A2 (en) 2010-10-19 2012-04-26 Trustees Of Tufts College Silk fibroin-based microneedles and methods of making the same
WO2012145739A1 (en) 2011-04-21 2012-10-26 Trustees Of Tufts College Compositions and methods for stabilization of active agents
EP2527368A1 (en) 2007-11-02 2012-11-28 Eli Lilly and Company Anti-hepcidin antibodies and uses thereof
WO2014145002A2 (en) 2013-03-15 2014-09-18 Kluge Jonathan A Low molecular weight silk compositions and stabilizing silk compositions
US8940690B2 (en) 2009-01-28 2015-01-27 National Institutes Of Health (Nih) Synthetic conjugates and uses thereof
US9427475B2 (en) 2013-10-04 2016-08-30 Merck Sharp & Dohme Corp. Glucose-responsive insulin conjugates
US11376329B2 (en) 2013-03-15 2022-07-05 Trustees Of Tufts College Low molecular weight silk compositions and stabilizing silk compositions

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2008011389A (en) * 2006-03-09 2009-01-26 Aplagen Gmbh Modified molecules which promote hematopoiesis.
JPWO2012133349A1 (en) 2011-03-25 2014-07-28 株式会社カネカ Protein for affinity separation matrix
WO2013170147A1 (en) * 2012-05-11 2013-11-14 Yale University Compounds useful for promoting protein degradation and methods using same
AU2013318928A1 (en) * 2012-09-21 2015-04-09 Kaneka Corporation Protein ligand for affinity isolation matrix
US9273093B2 (en) 2012-10-11 2016-03-01 Protagonist Therapeutics, Inc. α4β7 peptide dimer antagonists
EP4091624A1 (en) 2013-03-15 2022-11-23 Protagonist Therapeutics, Inc. Hepcidin analogues and uses thereof
GB201311891D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compound
GB201311888D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compounds
CA2949215C (en) 2014-05-16 2023-03-14 Protagonist Therapeutics, Inc. .alpha.4.beta.7 integrin thioether peptide antagonists
KR102482790B1 (en) 2014-07-17 2022-12-29 프로타고니스트 테라퓨틱스, 인코포레이티드 Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases
US10301371B2 (en) 2014-10-01 2019-05-28 Protagonist Therapeutics, Inc. Cyclic monomer and dimer peptides having integrin antagonist activity
MX2017004300A (en) 2014-10-01 2017-12-18 Protagonist Therapeutics Inc NOVEL A4ß7 PEPTIDE MONOMER AND DIMER ANTAGONISTS.
US10787490B2 (en) 2015-07-15 2020-09-29 Protaganist Therapeutics, Inc. Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
CA3009834A1 (en) 2015-12-30 2017-07-06 Protagonist Therapeutics, Inc. Analogues of hepcidin mimetics with improved in vivo half lives
US10407468B2 (en) 2016-03-23 2019-09-10 Protagonist Therapeutics, Inc. Methods for synthesizing α4β7 peptide antagonists
US10278957B2 (en) 2017-09-11 2019-05-07 Protagonist Therapeutics, Inc. Opioid agonist peptides and uses thereof
WO2019157268A1 (en) 2018-02-08 2019-08-15 Protagonist Therapeutics, Inc. Conjugated hepcidin mimetics
CN114341161A (en) 2019-07-10 2022-04-12 领导医疗有限公司 Peptide inhibitors of interleukin-23 receptor and their use for the treatment of inflammatory diseases
EP4090670A1 (en) 2020-01-15 2022-11-23 Janssen Biotech, Inc. Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
JP7397239B2 (en) 2020-11-20 2023-12-12 ヤンセン ファーマシューティカ エヌ.ベー. Compositions of peptide inhibitors of interleukin-23 receptors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5767078A (en) * 1995-06-07 1998-06-16 Johnson; Dana L. Agonist peptide dimers
US6077939A (en) * 1996-08-02 2000-06-20 Ortho-Mcneil Pharmaceutical, Inc. Methods and kits for making polypeptides having a single covalently bound N-terminal water-soluble polymer
US6221608B1 (en) * 1997-01-22 2001-04-24 Ortho Pharmaceutical Corporation Methods for identifying erythropoietin receptor binding protein

Family Cites Families (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
NZ210501A (en) * 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
KR850004274A (en) * 1983-12-13 1985-07-11 원본미기재 Method for preparing erythropoietin
US4677195A (en) * 1985-01-11 1987-06-30 Genetics Institute, Inc. Method for the purification of erythropoietin and erythropoietin compositions
US5006333A (en) * 1987-08-03 1991-04-09 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
US5080891A (en) * 1987-08-03 1992-01-14 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
WO1990008822A1 (en) * 1989-02-03 1990-08-09 Genetics Institute, Inc. Erythropoietin receptor
US5292654A (en) * 1990-12-13 1994-03-08 Whitehead Institute For Biomedical Research Mutant EPO receptor and uses therefor
US6153407A (en) * 1992-07-28 2000-11-28 Beth Israel Deaconess Medical Center Erythropoietin DNA having modified 5' and 3' sequences and its use to prepare EPO therapeutics
US5614184A (en) * 1992-07-28 1997-03-25 New England Deaconess Hospital Recombinant human erythropoietin mutants and therapeutic methods employing them
US5919455A (en) * 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
US5830851A (en) * 1993-11-19 1998-11-03 Affymax Technologies N.V. Methods of administering peptides that bind to the erythropoietin receptor
US5773569A (en) * 1993-11-19 1998-06-30 Affymax Technologies N.V. Compounds and peptides that bind to the erythropoietin receptor
US5580853A (en) * 1994-03-22 1996-12-03 New England Deaconess Hospital Modified polypeptides with increased biological activity
US5747446A (en) * 1994-03-22 1998-05-05 Beth Israel Deaconess Medical Center Modified polypeptides with increased biological activity
US5919758A (en) * 1994-03-22 1999-07-06 Beth Israel Deaconess Medical Center Modified polypeptides with altered biological activity
US5683983A (en) * 1995-06-07 1997-11-04 Glaxo Group Limited Peptides and compounds that bind to the IL-5 receptor
US5677280A (en) * 1995-06-07 1997-10-14 Glaxo Group Limited Peptides and compounds that bind to the IL-5 receptor
US5869451A (en) * 1995-06-07 1999-02-09 Glaxo Group Limited Peptides and compounds that bind to a receptor
US5668110A (en) * 1995-06-07 1997-09-16 Affymax Technologies N.V. Peptides and compounds that bind to the IL-5 receptor
US6251864B1 (en) * 1995-06-07 2001-06-26 Glaxo Group Limited Peptides and compounds that bind to a receptor
US5654276A (en) * 1995-06-07 1997-08-05 Affymax Technologies N.V. Peptides and compounds that bind to the IL-5 receptor
DK0885242T3 (en) * 1995-06-07 2008-07-14 Glaxo Group Ltd Peptides and compounds that bind to a thrombopoietin receptor
US5672662A (en) * 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
US6346390B1 (en) * 1996-03-08 2002-02-12 Receptron, Inc. Receptor derived peptides involved in modulation of response to ligand binding
US6103879A (en) * 1996-06-21 2000-08-15 Axys Pharmaceuticals, Inc. Bivalent molecules that form an activating complex with an erythropoietin receptor
US5932546A (en) * 1996-10-04 1999-08-03 Glaxo Wellcome Inc. Peptides and compounds that bind to the thrombopoietin receptor
US6103536A (en) * 1997-05-02 2000-08-15 Silver Lake Research Corporation Internally referenced competitive assays
ES2245114T3 (en) * 1998-08-06 2005-12-16 Mountain View Pharmaceuticals, Inc. CONJUGATES OF PEG-OXIDASA DE URATO AND ITS USE.
US6783965B1 (en) * 2000-02-10 2004-08-31 Mountain View Pharmaceuticals, Inc. Aggregate-free urate oxidase for preparation of non-immunogenic polymer conjugates
CN1330675A (en) * 1998-08-28 2002-01-09 格莱风科学公司 Polyamide chains of precise length, methods to manufacture them and their conjugates with protein
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
JP2002529502A (en) * 1998-11-17 2002-09-10 スミスクライン・ビーチャム・コーポレイション Treatment of thrombocytopenia
JO2291B1 (en) * 1999-07-02 2005-09-12 اف . هوفمان لاروش ايه جي Erythopintin derivatives
CZ299516B6 (en) * 1999-07-02 2008-08-20 F. Hoffmann-La Roche Ag Erythropoietin glycoprotein conjugate, process for its preparation and use and pharmaceutical composition containing thereof
AUPQ873300A0 (en) * 2000-07-12 2000-08-03 Medvet Science Pty. Ltd. A binding motif of a receptor (2)
US6552008B1 (en) * 1999-09-24 2003-04-22 Smithkline Beecham Corporation Thrombopoietin mimetics
AU7445900A (en) * 1999-09-27 2001-04-30 Chugai Seiyaku Kabushiki Kaisha Novel hemopoietin receptor protein, nr12
US6858630B2 (en) * 1999-12-06 2005-02-22 Smithkline Beecham Corporation Naphthimidazole derivatives and their use as thrombopoietin mimetics
AU2001281499B2 (en) * 2000-03-21 2007-01-04 Wisconsin Alumni Research Foundation Methods and reagents for regulation of cellular responses in biological systems
US6777387B2 (en) * 2000-03-31 2004-08-17 Enzon Pharmaceuticals, Inc. Terminally-branched polymeric linkers containing extension moieties and polymeric conjugates containing the same
WO2001087329A1 (en) * 2000-05-15 2001-11-22 F. Hoffmann-La Roche Ag Liquid pharmaceutical composition containing an erythropoietin derivate
HUP0303056A2 (en) * 2000-08-02 2003-12-29 Ortho-Mcneil Pharmaceutical, Inc. Improved anti-viral and anti-tumor chemotherapy by administration of erythropoeitin
RU2003109746A (en) * 2000-09-08 2005-01-27 Грифон Терапьютикс, Инк. (Us) SYNTHETIC PROTEINS STIMULATING ERYTHROPOESIS
CN100528235C (en) * 2000-12-20 2009-08-19 霍夫曼-拉罗奇有限公司 Conjugates of erythropoietin
US6531121B2 (en) * 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US7767643B2 (en) * 2000-12-29 2010-08-03 The Kenneth S. Warren Institute, Inc. Protection, restoration, and enhancement of erythropoietin-responsive cells, tissues and organs
FR2823220B1 (en) * 2001-04-04 2003-12-12 Genodyssee NOVEL ERYTHROPOIETIN (EPO) POLYNUCLEOTIDES AND POLYPEPTIDES
US20020169128A1 (en) * 2001-04-09 2002-11-14 Geroge Sigounas Erythropoietin ameliorates chemotherapy-induced toxicity in vivo
AU2002345938A1 (en) * 2001-06-28 2003-03-03 Mountain View Pharmaceuticals, Inc. Polymer stabilized proteinases
US6784154B2 (en) * 2001-11-01 2004-08-31 University Of Utah Research Foundation Method of use of erythropoietin to treat ischemic acute renal failure
US20050176627A1 (en) * 2002-09-09 2005-08-11 Anthony Cerami Long acting erythropoietins that maintain tissue protective activity of endogenous erythropoietin
US8129330B2 (en) * 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
TWI364295B (en) * 2002-12-26 2012-05-21 Mountain View Pharmaceuticals Polymer conjugates of cytokines, chemokines, growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
PL219741B1 (en) * 2002-12-26 2015-07-31 Mountain View Pharmaceuticals Polymer conjugates of interferon-beta with enhanced biological potency

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5767078A (en) * 1995-06-07 1998-06-16 Johnson; Dana L. Agonist peptide dimers
US6077939A (en) * 1996-08-02 2000-06-20 Ortho-Mcneil Pharmaceutical, Inc. Methods and kits for making polypeptides having a single covalently bound N-terminal water-soluble polymer
US6221608B1 (en) * 1997-01-22 2001-04-24 Ortho Pharmaceutical Corporation Methods for identifying erythropoietin receptor binding protein

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7858747B2 (en) 2005-08-30 2010-12-28 Novo Nordisk A/S Expression of proteins in E.coli
US8106154B2 (en) * 2007-01-31 2012-01-31 Affymax, Inc. Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
EP2118127A2 (en) * 2007-01-31 2009-11-18 Affymax, Inc. Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
JP2010520855A (en) * 2007-01-31 2010-06-17 アフィーマックス・インコーポレイテッド Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
EP2118127A4 (en) * 2007-01-31 2010-12-01 Affymax Inc Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
WO2008104513A1 (en) * 2007-03-01 2008-09-04 Novo Nordisk A/S Expression of proteins in e. coli
JP2010519894A (en) * 2007-03-01 2010-06-10 ノボ・ノルデイスク・エー/エス Protein expression in E. coli
EP2527368A1 (en) 2007-11-02 2012-11-28 Eli Lilly and Company Anti-hepcidin antibodies and uses thereof
AU2008341661B9 (en) * 2007-12-12 2013-08-15 Jiangsu Hansoh Pharmaceutical Group Co., Ltd. An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
RU2493168C2 (en) * 2007-12-12 2013-09-20 Цзянсу Хэнсох Фармасьютикал Ко., Лтд. Peptide derivative mimetic of erythropoetin and its pharmaceutical salts, their production and application
KR101646929B1 (en) * 2007-12-12 2016-08-09 지앙수 한소 파마슈티칼 코. 리미티드 An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
KR20110025731A (en) * 2007-12-12 2011-03-11 지앙수 한소 파마슈티칼 코. 리미티드 An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
US8642545B2 (en) 2007-12-12 2014-02-04 Jiangsu Hansoh Pharmaceutical Co., Ltd Erythropoietin mimetic peptide derivative and its pharmaceutical salts, the preparation and uses thereof
CN101675080B (en) * 2007-12-12 2012-07-11 江苏豪森药业股份有限公司 An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
TWI419706B (en) * 2007-12-12 2013-12-21 Jiangsu Hansoh Pharmaceutical Co Ltd Erythropoietin mimics derivatives, medcine salt and preparation thereof and their use
JP2011506350A (en) * 2007-12-12 2011-03-03 ジエンス ハンセン ファーマセウティカル カンパニー リミテッド Erythropoietin mimetic peptide derivatives and pharmaceutically acceptable salts thereof, their production and use
WO2009079910A1 (en) * 2007-12-12 2009-07-02 Jiangsu Hansen Pharmaceutical Co., Ltd. An erythropoietin mimetic peptide derivatives and its pharmaceutical salt, the preparation and uses thereof
US8906850B2 (en) * 2009-01-28 2014-12-09 Smartcells, Inc. Crystalline insulin-conjugates
US10398781B2 (en) 2009-01-28 2019-09-03 Smartcells, Inc. Conjugate based systems for controlled drug delivery
US20110301083A1 (en) * 2009-01-28 2011-12-08 Zion Todd C Conjugate based systems for controlled drug delivery
US9050370B2 (en) * 2009-01-28 2015-06-09 Smartcells, Inc. Conjugate based systems for controlled drug delivery
US20110281791A1 (en) * 2009-01-28 2011-11-17 Zion Todd C Crystalline insulin-conjugates
US8940690B2 (en) 2009-01-28 2015-01-27 National Institutes Of Health (Nih) Synthetic conjugates and uses thereof
WO2012054582A2 (en) 2010-10-19 2012-04-26 Trustees Of Tufts College Silk fibroin-based microneedles and methods of making the same
EP3495015A1 (en) 2010-10-19 2019-06-12 Trustees Of Tufts College Silk fibroin-based microneedles and methods of making the same
EP4218891A1 (en) 2010-10-19 2023-08-02 Trustees Of Tufts College Silk fibroin-based microneedles and methods of making the same
WO2012145739A1 (en) 2011-04-21 2012-10-26 Trustees Of Tufts College Compositions and methods for stabilization of active agents
WO2014145002A2 (en) 2013-03-15 2014-09-18 Kluge Jonathan A Low molecular weight silk compositions and stabilizing silk compositions
EP3412682A1 (en) 2013-03-15 2018-12-12 Trustees Of Tufts College Low molecular weight silk compositions and stabilizing silk compositions
US11376329B2 (en) 2013-03-15 2022-07-05 Trustees Of Tufts College Low molecular weight silk compositions and stabilizing silk compositions
EP4180448A1 (en) 2013-03-15 2023-05-17 Trustees of Tufts College Low molecular weight silk compositions and stabilizing silk compositions
US9427475B2 (en) 2013-10-04 2016-08-30 Merck Sharp & Dohme Corp. Glucose-responsive insulin conjugates
US9884125B2 (en) 2013-10-04 2018-02-06 Merck Sharp & Dohme Corp. Glucose-responsive insulin conjugates
US9889205B2 (en) 2013-10-04 2018-02-13 Merck Sharp & Dohme Corp. Glucose-responsive insulin conjugates

Also Published As

Publication number Publication date
US20080108564A1 (en) 2008-05-08
WO2006062685A3 (en) 2006-10-19
US20130012685A1 (en) 2013-01-10
US20110282029A1 (en) 2011-11-17

Similar Documents

Publication Publication Date Title
EP1625156B1 (en) Peptides that bind to the erythropoietin receptor
EP1629007B1 (en) Novel peptides that bind to the erythropoietin receptor
US20130012685A1 (en) Novel peptides that bind to the erythropoietin receptor
EP1917023A2 (en) Erythropoietin receptor peptide formulations and uses
US20120157660A1 (en) Novel peptides that bind to the erythropoietin receptor
AU2012203812A1 (en) Erythropoietin receptor peptide formulations and uses

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase

Ref document number: 05851594

Country of ref document: EP

Kind code of ref document: A2