US20040214996A1 - Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein - Google Patents

Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein Download PDF

Info

Publication number
US20040214996A1
US20040214996A1 US10/839,037 US83903704A US2004214996A1 US 20040214996 A1 US20040214996 A1 US 20040214996A1 US 83903704 A US83903704 A US 83903704A US 2004214996 A1 US2004214996 A1 US 2004214996A1
Authority
US
United States
Prior art keywords
residue
leu
nonfunctional
pth
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/839,037
Inventor
Paul Kostenuik
Chuan-Fa Liu
David Lacey
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen 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 Amgen Inc filed Critical Amgen Inc
Priority to US10/839,037 priority Critical patent/US20040214996A1/en
Publication of US20040214996A1 publication Critical patent/US20040214996A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K14/575Hormones
    • C07K14/635Parathyroid hormone (parathormone); Parathyroid hormone-related peptides
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • A61P5/16Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4 for decreasing, blocking or antagonising the activity of the thyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • A61P5/22Drugs for disorders of the endocrine system of the parathyroid hormones for decreasing, blocking or antagonising the activity of calcitonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/46Drugs for disorders of the endocrine system of the suprarenal hormones for decreasing, blocking or antagonising the activity of glucocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP) play important physiological roles in calcium homeostasis and in development, respectively.
  • Calcium concentration in the blood is tightly regulated, due to the essential role of calcium in cell metabolism.
  • PTH is an endocrine hormone which is secreted from the parathyroid gland in response to decreased serum calcium levels.
  • PTH acts directly to increase bone resorption and to stimulate renal calcium reabsorption, thus increasing or preserving circulating calcium stores.
  • PTH also indirectly increases calcium absorption in the gut by stimulating the renal hydroxylation of vitamin D.
  • Both primary and secondary hyperparathyroidism are conditions that are associated with excessive levels of circulating parathyroid hormone. Through the aforementioned pathways, excess PTH levels can cause hypercalcemia and osteopenia. Bone resorption inhibitors such as bisphosphonates and OPG can effectively protect bone and can inhibit the skeleton's contribution to hypercalcemia. However, the calcemic effects of hyperparathyroidism on the kidney and gut are not addressed by currently available therapy.
  • PTHrP is produced by many cell types, and plays an important role in regulating skeletal development. Postnatally, the roles for PTHrP are less clearly defined. Circulating levels of PTHrP are essentially non-detectable in normal healthy adults. However, many tumors of diverse embryological origins produce and secrete PTHrP in quantities sufficient to cause hypercalcemia. In fact, humoral hypercalcemia of malignancy (HHM) is the most common paraneoplastic syndrome, which accounts for significant patient morbidity and mortality.
  • HHM humoral hypercalcemia of malignancy
  • HHM is treated with saline hydration followed by bone resorption inhibitors such as bisphosphonates.
  • This treatment regimen typically takes 3-4 days to achieve significant reductions in serum calcium, and the effects are relatively short-lived (less than one month).
  • the effects of current treatment options are even less impressive.
  • Repeated administration of conventional therapies are usually progressively less effective.
  • N-terminal fragment of PTH has been used as a therapeutic agent. Intermittently administered native PTH-(1-84) exhibits osteogenic properties, and it has been recognized for decades that these properties can be fully realized with the C-terminally truncated fragment PTH-(1-34). Both peptides bind and activate the PTH-1 receptor with similar affinities, causing the activation of adenylate cyclase (AC) as well as phospholipase C (PLC). AC activation through PTH-1 receptor generates cAMP, while PLC activation through PTH-1 receptor generates PKC and intracellular calcium transients. PTH-(1-34) can maximally activate both the AC and the PLC pathways.
  • AC adenylate cyclase
  • PLC phospholipase C
  • Truncated PTH fragments have diminished AC/cAMP activation and similarly diminished anabolic activity.
  • Such truncated PTH fragments have this diminished activity (Rixon et al. (1994); Hilliker et al. (1996); Lane et al. (1996)) even if they maintain full agonism towards PKC. Rixon et al., (1994).
  • hPTH-(1-31) has a slightly reduced (1-6 fold) affinity for PTH-1 receptor compared to hPTH-(1-34), while hPTH-(1-30) has a significantly reduced (10-100 fold) affinity Takasu (1998). Perhaps due to this decreased PTH-1 receptor affinity, PTH-(1-30) is a weak and incomplete agonist for PLC activation via the rat PTH-1 receptor.
  • PTH-(1-31) Compared to PTH-(1-34), PTH-(1-31) has similar or slightly reduced anabolic potential (Rixon et al. (1994); Whitfield et al. (1996), Calcified Tissue International 53: 81-7; Whitfield et al. (1996), Calcified Tissue International 65: 143-7), binding affinity for PTH1R, and cAMP induction (Takasu (1998)). PTH-(1-31) also has slightly reduced PLC activation. Takasu (1998).
  • PTH-(1-31) and PTH-(1-34) had similar stimulatory effects on plasma and urinary cAMP concentration, but unlike PTH-(1-34), PTH-(1-31) failed to elevate serum calcium, plasma 1,25(OH)2D3, or urinary N-TX levels. Fraher et al. (1999), J. Clin. Endocrin. Met. 84: 2739-43. These data suggest that PTH-(1-31) has diminished capacity to induce bone resorption and to stimulation vitamin D synthesis, which is a favorable profile for bone anabolic agents.
  • PTH-(1-30) was initially shown to lack anabolic properties Whitfield et al. (1996), Calcified Tissue International 53: 81-7. More recently, however, it has been demonstrated that PTH-(1-30) is anabolic when administered at very high doses (400-2,000 ⁇ g/kg, vs. 80 ⁇ g/kg for PTH-(1-34)). The lower potency of PTH-(1-30) could be predicted by its lower binding affinity for PTH-1 receptor, its diminished cAMP activation, and/or to its greatly diminished PKC activation. Takasu (1998). It remains to be determined whether PTH-(1-30) has a similar or even more desirable reduction in apparent bone resorption activity.
  • PTH-(1-28) is the smallest reported fragment to fully activate cAMP. Neugebauer et al. (1995), Biochem. 34: 8835-42. However, hPTH-(1-28) was initially reported to have no osteogenic effects in OVX rats. Miller et al. (1997), J. Bone Min. Res. 12: S320 (Abstract). Recently, a very high dose of PTH-(1-28) (1,000 ⁇ g/kg/day) was shown to be anabolic in OVX rats, whereas 200 ⁇ g/kg/day was ineffective. Whitfield et al. (2000), J. Bone Min. Res. 15: 964-70. The diminished or absent anabolic effects of some truncated PTH fragments has been attributed to rapid clearance in vivo. Rixon et al. (1994).
  • Recombinant and modified proteins are an emerging class of therapeutic agents.
  • Useful modifications of protein therapeutic agents include combination with the “Fc” domain of an antibody and linkage to polymers such as polyethylene glycol (PEG) and dextran.
  • PEG polyethylene glycol
  • Such modifications are discussed in detail in a patent application entitled, “Modified Peptides as Therapeutic Agents,” U.S. Ser. No. 09/428,082, PCT appl. no. WO 99/25044, which is hereby incorporated by reference in its entirety.
  • a much different approach to development of therapeutic agents is peptide library screening.
  • the interaction of a protein ligand with its receptor often takes place at a relatively large interface.
  • the bulk of the protein ligand merely displays the binding epitopes in the right topology or serves functions unrelated to binding.
  • molecules of only “peptide” length (2 to 40 amino acids) can bind to the receptor protein of a given large protein ligand.
  • Such peptides may mimic the bioactivity of the large protein ligand (“peptide agonists”) or, through competitive binding, inhibit the bioactivity of the large protein ligand (“peptide antagonists”).
  • Phage display peptide libraries have emerged as a powerful method in identifying such peptide agonists and antagonists. See, for example, Scott et al. (1990), Science 249: 386; Devlin et al. (1990), Science 249: 404; U.S. Pat. No. 5,223,409, issued Jun. 29, 1993; U.S. Pat. No. 5,733,731, issued Mar. 31, 1998; U.S. Pat. No. 5,498,530, issued Mar. 12, 1996; U.S. Pat. No. 5,432,018, issued Jul. 11, 1995; U.S. Pat. No. 5,338,665, issued Aug. 16, 1994; U.S. Pat. No. 5,922,545, issued Jul.
  • random peptide sequences are displayed by fusion with coat proteins of filamentous phage.
  • the displayed peptides are affinity-eluted against an antibody-immobilized extracellular domain of a receptor.
  • the retained phages may be enriched by successive rounds of affinity purification and repropagation.
  • the best binding peptides may be sequenced to identify key residues within one or more structurally related families of peptides. See, e.g., Cwirla et al. (1997), Science 276: 1696-9, in which two distinct families were identified.
  • the peptide sequences may also suggest which residues may be safely replaced by alanine scanning or by mutagenesis at the DNA level. Mutagenesis libraries may be created and screened to further optimize the sequence of the best binders. Lowman (1997), Ann. Rev. Biophys. Biomol. Struct. 26: 401-24.
  • Structural analysis of protein-protein interaction may also be used to suggest peptides that mimic the binding activity of large protein ligands.
  • the crystal structure may suggest the identity and relative orientation of critical residues of the large protein ligand, from which a peptide may be designed. See, e.g., Takasaki et al. (1997), Nature Biotech. 15: 1266-70.
  • These analytical methods may also be used to investigate the interaction between a receptor protein and peptides selected by phage display, which may suggest further modification of the peptides to increase binding affinity.
  • E. coli display Other methods compete with phage display in peptide research.
  • a peptide library can be fused to the carboxyl terminus of the lac repressor and expressed in E. coli.
  • Another E. coli -based method allows display on the cell's outer membrane by fusion with a peptidoglycan-associated lipoprotein (PAL).
  • PAL peptidoglycan-associated lipoprotein
  • these and related methods are collectively referred to as “ E. coli display.”
  • translation of random RNA is halted prior to ribosome release, resulting in a library of polypeptides with their associated RNA still attached.
  • RNA-peptide screening Chemically derived peptide libraries have been developed in which peptides are immobilized on stable, non-biological materials, such as polyethylene rods or solvent-permeable resins. Another chemically derived peptide library uses photolithography to scan peptides immobilized on glass slides.
  • Chemical-peptide screening may be advantageous in that it allows use of D-amino acids and other unnatural analogues, as well as non-peptide elements. Both biological and chemical methods are reviewed in Wells & Lowman (1992), Curr. Opin. Biotechnol. 3: 355-62. Conceptually, one may discover peptide mimetics of any protein using phage display, RNA-peptide screening, and the other methods mentioned above.
  • modulators of PTH and PTHrP comprise:
  • a PTH/PTHrP modulating domain preferably the amino acid sequence of PTH/PTHrP modulating domains of PTH and/or PTHrP, or sequences derived therefrom by phage display, RNA-peptide screening, or the other techniques mentioned above;
  • a vehicle such as a polymer (e.g., PEG or dextran) or an Fc domain, which is preferred;
  • PTH/PTHrP modulating domains comprise the PTH and PTHrP-derived amino acid sequences described hereinafter.
  • Other PTH/PTHrP modulating domains can be generated by phage display, RNA-peptide screening and the other techniques mentioned herein. Such peptides typically will be antagonists of both PTH and PTHrP, although such techniques can be used to generate peptide sequences that serve as selective inhibitors (e.g., inhibitors of PTH but not PTHrP).
  • Step (a) is preferably carried out by selection from the peptide sequences in Tables 1 and 2 hereinafter or from phage display, RNA-peptide screening, or the other techniques mentioned herein.
  • the compounds of this invention may be prepared by standard synthetic methods, recombinant DNA techniques, or any other methods of preparing peptides and fusion proteins.
  • Compounds of this invention that encompass non-peptide portions may be synthesized by standard organic chemistry reactions, in addition to standard peptide chemistry reactions when applicable.
  • the primary use contemplated for the compounds of this invention is as therapeutic or prophylactic agents.
  • the vehicle-linked peptide may have activity comparable to—or even greater than—the natural ligand mimicked by the peptide.
  • the compounds of this invention may be used for therapeutic or prophylactic purposes by formulating them with appropriate pharmaceutical carrier materials and administering an effective amount to a patient, such as a human (or other mammal) in need thereof.
  • a patient such as a human (or other mammal) in need thereof.
  • Other related aspects are also included in the instant invention.
  • PTH-(1-28) or (1-34) molecules comprising PTH/PTHRP modulating domains having a shortened PTH C-terminal sequence
  • PTH-(1-28) or (1-34) shows no anabolic studies using sustained duration delivery of such C-terminally truncated PTH fragments.
  • molecules comprising smaller fragments such as PTH-(1-30)-Fc can be anabolic on their own.
  • hPTH-(1-30) is nearly as effective at cAMP stimulation as is hPTH-(1-34).
  • PTH fragments may be selectively related to their cAMP activation, rather than PLC activation, so that PTH fragments with reduced receptor affinity will have a favorable anabolic profile. It is possible that continuous exposure to truncated PTH fragments would have a different, and more favorable effect on bone compared to continuous exposure to PTH-(1-34) or PTH-(1-84) that has been demonstrated in humans by Fraher et al. (1999).
  • FIG. 1 shows exemplary Fc dimers that may be derived from an IgG1 antibody.
  • Fc in the figure represents any of the Fc variants within the meaning of “Fc domain” herein.
  • X 1 ” and “X 2 ” represent peptides or linker-peptide combinations as defined hereinafter. The specific dimers are as follows:
  • A Single disulfide-bonded dimers.
  • IgG1 antibodies typically have two disulfide bonds at the hinge region between the constant and variable domains.
  • the Fc domain in FIG. 1A may be formed by truncation between the two disulfide bond sites or by substitution of a cysteinyl residue with an unreactive residue (e.g., alanyl).
  • the Fc domain is linked at the C-terminus of the peptide.
  • B Doubly disulfide-bonded dimers.
  • This Fc domain may be formed by truncation of the parent antibody to retain both cysteinyl residues in the Fc domain chains or by expression from a construct including a sequence encoding such an Fc domain.
  • the Fc domain is linked at the C-terminus of the peptide.
  • C Noncovalent dimers.
  • This Fc domain may be formed by elimination of the cysteinyl residues by either truncation or substitution. One may desire to eliminate the cysteinyl residues to avoid impurities formed by reaction of the cysteinyl residue with cysteinyl residues of other proteins present in the host cell. The noncovalent bonding of the Fc domains is sufficient to hold together the dimer.
  • Other dimers may be formed by using Fc domains derived from different types of antibodies (e.g., IgG2, IgM).
  • FIG. 2 shows the structure of additional compounds of the invention.
  • FIG. 2A shows a single chain molecule and may also represent the DNA construct for the molecule.
  • FIG. 2B shows a dimer in which the linker-peptide portion is present on only one chain of the dimer.
  • FIG. 2C shows a dimer having the peptide portion on both chains. The dimer of FIG. 2C will form spontaneously in certain host cells upon expression of a DNA construct encoding the single chain as shown in FIG. 3. In other host cells, the cells could be placed in conditions favoring formation of dimers or the dimers can be formed in vitro.
  • FIG. 3 shows exemplary nucleic acid and amino acid sequences (SEQ ID NOS: 1 and 2, respectively) of human IgG1 Fc that may be used in this invention.
  • FIG. 4 shows the calcemic response of normal mice to PTH-(1-34) and to PTH-(1-34)-Fc.
  • Mice were challenged with vehicle (PBS, —X—), or with PTH-(1-34) (open symbols) or with PTH-(1-34)-Fc (closed symbols).
  • FIG. 5 shows that [Asn10,Leu11]PTHrP-(7-34)-Fc inhibits the calcemic response of normal mice to PTHrP.
  • Normal male mice were injected SC with vehicle (PBS, circles) or with human PTHrP-(1-34) at 0.5 mg/kg (squares).
  • PTHrP-challenged mice were then immediately injected SC with [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 mg/kg (triangles) or 30 mg/kg (diamonds).
  • Data represent group means, with an n of 6 mice/group.
  • FIG. 6 shows the effect of [Asn10,Leu11]PTHrP-(7-34)-Fc on chronic hypercalcemia induced by PTH-(1-34)-Fc.
  • Normal male mice were challenged once by SC injection with PTH-(1-34)-Fc (30 mg/kg) (open circles), or with vehicle (PBS, open squares).
  • Some PTH-(1-34)-Fc-challenged mice were treated once, at the time of challenge, with [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 (closed triangle), 30 (closed circle), or 100 mg/kg (closed square). All doses of [Asn10,Leu11]PTHrP-(7-34)-Fc caused a significant suppression of PTH-(1-34)-Fc-mediated hypercalcemia.
  • FIG. 7 shows cAMP accumulation in ROS 17/2.8 rat osteoblast-like cells. Cultures were treated with the phosphodiesterase inhibitor IBMX and then challenged for 15 minutes with various PTH fragments. cAMP was measured by ELISA.
  • FIG. 8 shows the effects of single treatments on clinical chemistry. Peripheral blood was obtained daily for 3 days following single subcutaneous injections of the indicated compounds.
  • FIG. 8A shows total serum calcium
  • FIG. 8B alkaline phosphatase (AP), a marker of osteoblast activity
  • FIG. 8C tartrate-resistant acid phosphatase (TRAP), a marker of osteoclast activity
  • FIG. 8D AP:TRAP ratio, an index of relative osteoblas: osteoclast activity.
  • FIG. 9 shows the effects of PTH constructs on bone mineral density.
  • Peripheral quantitative computed tomography pQCT was performed on the proximal tibial metaphysis of mice on day 15, after injections of PTH constructs on day 0, 5 and 10.
  • FIG. 10 shows the effect of twice-weekly PTH-(1-34)-Fc versus daily PTH-(1-34) on tibial, trabecular, and cortical bone mineral density (BMD).
  • Daily PTH [PTH-(1-34)] was given at 80 ⁇ g/kg/day (20 nmol/kg/day).
  • FIG. 11 shows the effects of twice-weekly treatment on BMD and serum calcium in aged ovariectomized (OVX) rats. Eleven months after OVX, rats were treated twice per week with phosphate-buffered saline (PBS, vehicle) or with APD (0.5 mg/kg) or with PTH-(1-34)-Fc (50 nmol/kg). DEXA was performed weekly. Blood was drawn 24 hours after the second weekly injection, when the calcemic effects of PTH-Fc are typically maximal.
  • PBS phosphate-buffered saline
  • APD 0.5 mg/kg
  • PTH-(1-34)-Fc 50 nmol/kg
  • FIG. 12 shows the effect of a single subcutaneous injection of PTH-(1-34)-Fc into OVX cynomologus monkeys.
  • the dotted line indicates the threshold for hypercalcemia, based on an elevation of calcium greater than three standard deviations above the normal mean, on two or more consecutive timepoints.
  • a compound may include additional amino acids on either or both of the N— or C-termini of the given sequence. Of course, these additional amino acids should not significantly interfere with the activity of the compound.
  • amino residue refers to amino acid residues in D- or L-form having sidechains comprising acidic groups.
  • Exemplary acidic residues include D and E.
  • aromatic residue refers to amino acid residues in D- or L-form having sidechains comprising aromatic groups.
  • exemplary aromatic residues include F, Y, and W.
  • basic residue refers to amino acid residues in D- or L-form having sidechains comprising basic groups.
  • Exemplary basic residues include H, K, and R.
  • hydrophilic residue and “Haa” refer to amino acid residues in D- or L-form having sidechains comprising at least one hydrophilic functional group or polar group.
  • exemplary hydrophilic residues include C, D, E, H, K, N, Q, R, S, and T.
  • lipophilic residue and “Laa” refer to amino acid residues in D- or L-form having sidechains comprising uncharged, aliphatic or aromatic groups.
  • Exemplary lipophilic sidechains include F, I, L, M, V, W, and Y.
  • Alanine (A) is amphiphilic—it is capable of acting as a hydrophilic or lipophilic residue. Alanine, therefore, is included within the definition of both “lipophilic residue” and “hydrophilic residue.”
  • nonfunctional residue refers to amino acid residues in D- or L-form having sidechains that lack acidic, basic, or aromatic groups.
  • exemplary nonfunctional amino acid residues include M, G, A, V, I, L and norleucine (Nle).
  • vehicle refers to a molecule that prevents degradation and/or increases half-life, reduces toxicity, reduces immunogenicity, or increases biological activity of a therapeutic protein.
  • exemplary vehicles include an Fc domain (which is preferred) as well as a linear polymer (e.g., polyethylene glycol (PEG), polylysine, dextran, etc.); a branched-chain polymer (see, for example, U.S. Pat. No. 4,289,872 to Denkenwalter et al., issued Sep. 15, 1981; U.S. Pat. No. 5,229,490 to Tam, issued Jul. 20, 1993; WO 93/21259 by Frechet et al., published 28 Oct.
  • lipid a lipid
  • a cholesterol group such as a steroid
  • a carbohydrate or oligosaccharide e.g., dextran
  • HSA human serum albumin
  • TTR transtheratin
  • Vehicles are further described hereinafter.
  • native Fc refers to molecule or sequence comprising the sequence of a non-antigen-binding fragment resulting from digestion of whole antibody, whether in monomeric or multimeric form.
  • the original immunoglobulin source of the native Fc is preferably of human origin and may be any of the immunoglobulins, although IgG1 and IgG2 are preferred.
  • Native Fc's are made up of monomeric polypeptides that may be linked into dimeric or multimeric forms by covalent (i.e., disulfide bonds) and non-covalent association.
  • the number of intermolecular disulfide bonds between monomeric subunits of native Fc molecules ranges from 1 to 4 depending on class (e.g., IgG, IgA, IgE) or subclass (e.g., IgG1, IgG2, IgG3, IgA1, IgGA2).
  • class e.g., IgG, IgA, IgE
  • subclass e.g., IgG1, IgG2, IgG3, IgA1, IgGA2
  • One example of a native Fc is a disulfide-bonded dimer resulting from papain digestion of an IgG (see Ellison et al. (1982), Nucleic Acids Res. 10: 4071-9).
  • native Fc as used herein is generic to the monomeric, dimeric, and multimeric forms.
  • Fc variant refers to a molecule or sequence that is modified from a native Fc but still comprises a binding site for the salvage receptor, FcRn.
  • International applications WO 97/34631 published 25 Sep. 1997) and WO 96/32478 describe exemplary Fc variants, as well as interaction with the salvage receptor, and are hereby incorporated by reference in their entirety.
  • the term “Fc variant” comprises a molecule or sequence that is humanized from a non-human native Fc.
  • a native Fc comprises sites that may be removed because they provide structural features or biological activity that are not required for the fusion molecules of the present invention.
  • Fc variant comprises a molecule or sequence that lacks one or more native Fc sites or residues that affect or are involved in (1) disulfide bond formation, (2) incompatibility with a selected host cell (3) N-terminal heterogeneity upon expression in a selected host cell, (4) glycosylation, (5) interaction with complement, (6) binding to an Fc receptor other than a salvage receptor, or (7) antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • Fc domain encompasses native Fc and Fc variant molecules and sequences as defined above. As with Fc variants and native Fc's, the term “Fc domain” includes molecules in monomeric or multimeric form, whether digested from whole antibody or produced by other means.
  • multimer as applied to Fc domains or molecules comprising Fc domains refers to molecules having two or more polypeptide chains associated covalently, noncovalently, or by both covalent and non-covalent interactions.
  • IgG molecules typically form dimers; IgM, pentamers; IgD, dimers; and IgA, monomers, dimers, trimers, or tetramers. Multimers may be formed by exploiting the sequence and resulting activity of the native Ig source of the Fc or by derivatizing (as defined below) such a native Fc.
  • dimers as applied to Fc domains or molecules comprising Fc domains refers to molecules having two polypeptide chains associated covalently or non-covalently.
  • exemplary dimers within the scope of this invention are as shown in FIGS. 1 and 2.
  • the terms “derivatizing” and “derivative” or “derivatized” comprise processes and resulting compounds respectively in which (1) the compound has a cyclic portion; for example, cross-linking between cysteinyl residues within the compound; (2) the compound is cross-linked or has a cross-linking site; for example, the compound has a cysteinyl residue and thus forms cross-linked dimers in culture or in vivo; (3) one or more peptidyl linkage is replaced by a non-peptidyl linkage; (4) the N-terminus is replaced by —NRR 1 , NRC(O)R 1 , —NRC(O)OR 1 , —NRS(O) 2 R 1 , —NHC(O)NHR, a succinimide group, or substituted or unsubstituted benzyloxycarbonyl-NH—, wherein R and R 1 and the ring substituents are as defined hereinafter; (5) the C-terminus is replaced by —C(
  • peptide refers to molecules of 1 to 85 amino acids, with molecules of 5 to 34 amino acids preferred.
  • exemplary peptides may comprise the PTH/PTHrP modulating domain of a naturally occurring molecule or comprise randomized sequences.
  • randomized refers to fully random sequences (e.g., selected by phage display methods or RNA-peptide screening) and sequences in which one or more residues of a naturally occurring molecule is replaced by an amino acid residue not appearing in that position in the naturally occurring molecule.
  • Exemplary methods for identifying peptide sequences include phage display, E. coli display, ribosome display, RNA-peptide screening, chemical screening, and the like.
  • PTH/PTHrP modulating domain refers to any amino acid sequence that binds to the PTH-1 receptor and/or the PTH-2 receptor and comprises naturally occurring sequences or randomized sequences. Exemplary PTH/PTHrP modulating domains can be identified or derived as described in the references listed for Tables 1 and 2, which are hereby incorporated by reference in their entirety.
  • PTH agonist refers to a molecule that binds to PTH-1 or PTH-2 receptor and increases or decreases one or more PTH activity assay parameters as does full-length native human parathyroid hormone.
  • An exemplary PTH activity assay is disclosed in Example 1.
  • PTH antagonist refers to a molecule that binds to PTH-1 or PTH-2 receptor and blocks or prevents the normal effect on those parameters by full length native human parathyroid hormone.
  • An exemplary PTH activity assay is disclosed in Example 2.
  • bone resorption inhibitor refers to such molecules as determined by the assays of Examples 4 and 11 of WO 97/23614:, which is hereby incorporated by reference in its entirety.
  • Exemplary bone resorption inhibitors include OPG and OPG-L antibody, which are described in WO 97/23614 and WO98/46751, respectively, which are hereby incorporated by reference in their entirety.
  • physiologically acceptable salts of the compounds of this invention are also encompassed herein.
  • physiologically acceptable salts is meant any salts that are known or later discovered to be pharmaceutically acceptable. Some specific examples are: acetate; trifluoroacetate; hydrohalides, such as hydrochloride and hydrobromide; sulfate; citrate; tartrate; glycolate; and oxalate.
  • the present inventors identified in particular preferred sequences derived from PTH and PTHrP. These sequences can be randomized through the techniques mentioned above by which one or more amino acids may be changed while maintaining or even improving the binding affinity of the peptide.
  • the peptide may be attached to the vehicle through the peptide's C-terminus.
  • vehicle-peptide molecules of this invention may be described by the following formula I:
  • F 1 is a vehicle (preferably an Fc domain) and is attached at the C-terminus of P 1 -(L 1 ) a ;
  • P 1 is a sequence of a PTH/PTHrP modulating domain
  • L 1 is a linker
  • a is 0 or 1.
  • Peptides Any number of peptides may be used in conjunction with the present invention. Peptides may comprise part of the sequence of naturally occurring proteins, may be randomized sequences derived from the sequence of the naturally occurring proteins, or may be wholly randomized sequences. Phage display and RNA-peptide screening, in particular, are useful in generating peptides for use in the present invention.
  • a PTH/PTHrP modulating domain sequence particularly of interest is of the formula II X N X 8 HX 10 X 11 X 12 KX 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 3) RX 21 X 22 X 23 X 24 X 25 X 26 X 27 X 28 X C
  • X N is absent or is X 3 X 4 X 5 X 6 X 7 , X 2 X 3 X 4 X 5 X 6 X 7 , X 1 X 2 X 3 X 4 X 5 X 6 X 7 , or YX 1 X 2 X 3 X 4 X 5 X 6 X 7 ;
  • X 1 is an amino acid residue (nonfunctional, hydrophilic or aromatic residue preferred; A, S or Y preferred);
  • X 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • X 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • X 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • X 5 is an amino acid residue (nonfunctional or basic residue preferred, H or I most preferred);
  • X 6 is an amino acid residue (acidic or hydrophilic residue preferred, Q or E most preferred);
  • X 7 is an amino acid residue (nonfunctional or aromatic residue preferred, L or F most preferred);
  • X 8 is an amino acid residue (nonfunctional residue preferred, M or Nle most preferred);
  • X 10 is an amino acid residue (an acidic or hydrophilic residue preferred, N or D most preferred);
  • X 11 is an amino acid residue (nonfunctional or basic residue preferred, L, R, or K most preferred);
  • X 12 is an amino acid residue (nonfunctional or aromatic residue preferred, G, F, or W most preferred);
  • X 14 is an amino acid residue (basic or hydrophilic residue preferred, H or S most preferred);
  • X 15 is an amino acid residue (nonfunctional residue preferred, with L or I most preferred);
  • X 16 is an amino acid residue (nonfunctional or hydrophilic residue preferred, Q, N, S, or A most preferred);
  • X 17 is an amino acid residue (acidic, hydrophilic, or nonfunctional residue preferred; S, D, or L most preferred);
  • X 18 is an amino acid residue (nonfunctional residue preferred, M, L, V or Nle most preferred);
  • X 19 is an amino acid residue (acidic or basic residue preferred, E or R most preferred);
  • X 21 is an amino acid residue (nonfunctional residue or basic residue preferred; V, M, R, or Nle most preferred);
  • X 22 is an amino acid residue (hydrophilic, acidic, or aromatic residue preferred, E or F most preferred);
  • X 23 is an aromatic or lipophilic residue (W or F preferred);
  • X 24 is a lipophilic residue (L preferred);
  • X 25 is an amino acid residue (hydrophilic or basic residue preferred, R or H most preferred);
  • X 26 is an amino acid residue (hydrophilic or basic residue preferred, K or H most preferred);
  • X 27 is an amino acid residue (lipophilic, basic, or nonfunctional residue preferred, K or L most preferred);
  • X 28 is an amino acid residue (lipophilic or nonfunctional residue preferred, L or I most preferred);
  • X C is absent or is X 29 , X 29 X 30 , X 29 X 30 X 31 , X 29 X 30 X 31 X 32 , X 29 X 30 X 31 X 32 X 33 , X 29 X 30 X 31 X 32 X 33 X 34 , X 29 X 30 X 31 X 32 X 33 X 34 X 35 , or X 29 X 30 X 31 X 32 X 33 X 34 X 35 X 36 ;
  • X 29 is an amino acid residue (hydrophilic or nonfunctional residue preferred, Q or A most preferred);
  • X 30 is an amino acid residue (hydrophilic or acidic residue preferred, D or E most preferred);
  • X 31 is an amino acid residue (lipophilic or nonfunctional residue preferred, V or I most preferred);
  • X 32 is an amino acid residue (basic residue preferred, H most preferred);
  • X 33 is an amino acid residue (hydrophilic residue preferred, N or T most preferred);
  • X 34 is an amino acid residue (nonfunctional or aromatic residue preferred, A, F or Y most preferred);
  • X 35 is an amino acid residue (acidic residue preferred, E most preferred).
  • X 36 is an amino acid residue (aromatic residue preferred, Y most preferred).
  • a preferred PTH/PTHrP modulating domain sequence formula is III J N J 7 J 8 HNLJ 12 KHLJ 16 SJ 18 (SEQ ID NO: 4) J 19 RJ 21 EWLRKKLJ C
  • J N is absent or is selected from J 1 J 2 J 3 J 4 J 5 J 6 , J 2 J 3 J 4 J 5 J 6 , J 3 J 4 J 5 J 6 ;
  • J 1 is an amino acid residue (nonfunctional, hydrophilic, or aromatic residue preferred; A, S or Y most preferred);
  • J 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • J 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • J 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • J 5 is an amino acid residue (nonfunctional residue preferred, I most preferred);
  • J 6 is an amino acid residue (basic residue preferred, Q preferred);
  • J 7 is an amino acid residue (nonfunctional or aromatic residue preferred, L or F most preferred);
  • J 8 is an amino acid residue (nonfunctional residue preferred, M or Nle most preferred);
  • J 12 is an amino acid residue (nonfunctional or aromatic residue preferred, G or W most preferred);
  • J 16 is an amino acid residue (nonfunctional or hydrophilic residue preferred, N, S, or A most preferred);
  • J 18 is an amino acid residue (nonfunctional residue preferred, M, Nle, L, or V most preferred);
  • J 19 is an acidic or basic residue (E or R preferred);
  • J 21 is an amino acid residue (nonfunctional residue preferred, V, M, or Nle most preferred);
  • J C is absent or is J 29 , J 29 J 30 , J 29 J 30 J 31 , J 29 J 30 J 31 J 32 , J 29 J 30 J 31 J 32 J 33 , or J 29 J 30 J 31 J 32 J 33 J 34 ;
  • J 29 is an amino acid residue (hydrophilic or nonfunctional residue preferred, Q or A most preferred);
  • J 30 is an amino acid residue (hydrophilic or acidic residue preferred, D or E most preferred);
  • J 31 is an amino acid residue (lipophilic or nonfunctional residue preferred, V or I most preferred);
  • J 32 is an amino acid residue (basic residue preferred, H most preferred);
  • J 33 is an amino acid residue (acidic residue preferred, N most preferred);
  • J 34 is an amino acid residue (aromatic residue preferred, F or Y most preferred).
  • PTH/PTHrP modulating domain sequence is IV O N LHO 10 O 11 O 12 KSIO 15 (SEQ ID NO: 5) O 16 LRRRFO 23 LHHLIO C
  • O N is absent or is YO 1 O 2 O 3 O 4 O 5 O 6 O 7 , O 1 O 2 O 3 O 4 O 5 O 6 O 7 , O 2 O 3 O 4 O 5 O 6 O 7 , O 3 O 4 O 5 O 6 O 7 , O 4 O 5 O 6 O 7 , O 5 6 O 7 , O 6 O 7 , or O 7 ;
  • O 1 is an amino acid residue (nonfunctional residue preferred, A most preferred);
  • O 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • O 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • O 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • O 5 is an amino acid residue (basic or nonfunctional residue preferred, H or I preferred);
  • O 6 is an amino acid residue (hydrophilic residue preferred, Q most preferred);
  • O 7 is an amino acid residue (nonfunctional residue preferred, L most preferred);
  • O 10 is an amino acid residue (acidic or hydrophilic residue preferred, N or D most preferred);
  • O 11 is an amino acid residue (basic or nonfunctional residue preferred, K or L most preferred);
  • O 12 is an amino acid residue (aromatic or nonfunctional residue preferred, G, F, or W most preferred);
  • O 15 is an amino acid residue (hydrophilic or nonfunctional residue preferred, I or S most preferred);
  • O 16 is an amino acid residue (hydrophilic residue preferred, Q or N most preferred);
  • O 17 is an amino acid residue (acidic or nonfunctional residue preferred, D or L most preferred);
  • O 23 is an amino acid residue (aromatic residue preferred, with F or W most preferred);
  • O C is absent or is O 29 , O 29 O 30 , O 29 O 30 O 31 , O 29 O 30 O 31 O 32 , O 29 O 30 O 31 O 32 O 33 , O 29 O 30 O 31 O 32 O 33 O 34 , O 29 O 30 O 31 O 32 O 33 O 34 O 35 , or O 29 O 30 O 31 O 32 O 33 O 34 O 35 O 36 ; and
  • O 29 through O 36 are each independently amino acid residues.
  • Exemplary peptide sequences for this invention appear in Tables 1 and 2 below. These peptides may be prepared as described in the cited references or in U.S. Pat. Nos. 4,423,037, 4,968,669, 5,001,22, and 6,051,686, each of which is hereby incorporated by reference in its entirety. Molecules of this invention incorporating these peptide sequences may be prepared by methods known in the art. Single letter amino acid abbreviations are used. Any of these peptides may be linked in tandem (i.e., sequentially), with or without linkers. Any peptide containing a cysteinyl residue may be cross-linked with another Cys-containing peptide, either or both of which may be linked to a vehicle.
  • PTH/PTHrP modulating domain has the sequence of the peptide known as TIP39: SLALADDAAFRERARLLAAL (SEQ ID NO: 160) ERRHWLNSYMHKLLVLDAP
  • TIP39 is described by Usdin et al. (1999), Nature Neurosci. 2(11): 941-3; Usdin et al. (1996), Endocrinology 137(10): 4285-97; Usdin et al. (1995), J. Biol. Chem. 270(26): 15455-8; Usdin et al. (1999), Endocrinol. 140(7): 3363-71.
  • Additional useful PTH/PTHrP modulating domain sequences may result from conservative and/or non-conservative modifications of the amino acid sequences of SEQ ID NOS: 3, 4, 5, TIP39, or the sequences listed in Tables 1 and 2.
  • Conservative modifications will produce peptides having functional and chemical characteristics similar to those of the PTH or PTHrP peptide from which such modifications are made.
  • substantial modifications in the functional and/or chemical characteristics of the peptides may be accomplished by selecting substitutions in the amino acid sequence that differ significantly in their effect on maintaining (a) the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the size of the molecule.
  • a “conservative amino acid substitution” may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position.
  • any native residue in the polypeptide may also be substituted with alanine, as has been previously described for “alanine scanning mutagenesis” (see, for example, MacLennan et al., 1998, Acta Physiol. Scand. Suppl. 643:55-67; Sasaki et al., 1998, Adv. Biophys. 35:1-24, which discuss alanine scanning mutagenesis).
  • Desired amino acid substitutions can be determined by those skilled in the art at the time such substitutions are desired.
  • amino acid substitutions can be used to identify important residues of the peptide sequence, or to increase or decrease the affinity of the peptide or vehicle-peptide molecules (see preceding formulae) described herein.
  • Exemplary amino acid substitutions are set forth in Table 3.
  • conservative amino acid substitutions also encompass non-naturally occurring amino acid residues which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems.
  • residues may be divided into classes based on common sidechain properties that may be useful for modifications of sequence. For example, non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues may be introduced into regions of the peptide that are homologous with non-human orthologs, or into the non-homologous regions of the molecule. In addition, one may also make modifications using P or G for the purpose of influencing chain orientation.
  • hydropathic index of amino acids may be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine ( ⁇ 0.4); threonine ( ⁇ 0.7); serine ( ⁇ 0.8); tryptophan ( ⁇ 0.9); tyrosine ( ⁇ 1.3); proline ( ⁇ 1.6); histidine ( ⁇ 3.2); glutamate ( ⁇ 3.5); glutamine ( ⁇ 3.5); aspartate ( ⁇ 3.5); asparagine ( ⁇ 3.5); lysine ( ⁇ 3.9); and arginine ( ⁇ 4.5).
  • hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine ( ⁇ 0.4); proline ( ⁇ 0.5 ⁇ 1); alanine ( ⁇ 0.5); histidine ( ⁇ 0.5); cysteine ( ⁇ 1.0); methionine ( ⁇ 1.3); valine ( ⁇ 1.5); leucine ( ⁇ 1.8); isoleucine ( ⁇ 1.8); tyrosine ( ⁇ 2.3); phenylalanine ( ⁇ 2.5); tryptophan ( ⁇ 3.4).
  • a skilled artisan will be able to determine suitable variants of the polypeptide as set forth in the foregoing sequences using well known techniques. For identifying suitable areas of the molecule that may be changed without destroying activity, one skilled in the art may target areas not believed to be important for activity. For example, when similar polypeptides with similar activities from the same species or from other species are known, one skilled in the art may compare the amino acid sequence of a peptide to similar peptides. With such a comparison, one can identify residues and portions of the molecules that are conserved among similar polypeptides.
  • One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar polypeptides. In view of that information, one skilled in the art may predict the alignment of amino acid residues of a peptide with respect to its three dimensional structure. One skilled in the art may choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art may generate test variants containing a single amino acid substitution at each desired amino acid residue. The variants can then be screened using activity assays know to those skilled in the art. Such data could be used to gather information about suitable variants.
  • One method of predicting secondary structure is based upon homology modeling. For example, two polypeptides or proteins which have a sequence identity of greater than 30%, or similarity greater than 40% often have similar structural topologies.
  • the recent growth of the protein structural data base (PDB) has provided enhanced predictability of secondary structure, including the potential number of folds within a polypeptide's or protein's structure. See Holm et al., Nucl. Acid. Res., 27(1): 244-247 (1999). It has been suggested (Brenner et al., Curr. Op. Struct. Biol., 7(3): 369-376 (1997)) that there are a limited number of folds in a given polypeptide or protein and that once a critical number of structures have been resolved, structural prediction will gain dramatically in accuracy.
  • Additional methods of predicting secondary structure include “threading” (Jones, D., Curr. Opin. Struct. Biol., 7(3): 377-87 (1997); Sippl et al., Structure, 4(1): 15-9 (1996)), “profile analysis” (Bowie et al., Science, 253: 164-170 (1991); Gribskov et al., Meth. Enzym., 183: 146-159 (1990); Gribskov et al., Proc. Nat. Acad. Sci., 84(13): 4355-8 (1987)), and “evolutionary linkage” (See Home, supra, and Brenner, supra).
  • Vehicles This invention requires the presence of at least one vehicle (F 1 ) attached to a peptide through the C-terminus or a sidechain of one of the amino acid residues.
  • F 1 vehicle
  • Multiple vehicles may also be used; e.g., an Fc at the C-terminus and a PEG group at a sidechain.
  • Fc domain is the preferred vehicle.
  • the Fc domain may be fused to the C terminus of the peptides.
  • Fc variants are suitable vehicles within the scope of this invention.
  • a native Fc may be extensively modified to form an Fc variant in accordance with this invention, provided binding to the salvage receptor is maintained; see, for example WO 97/34631 and WO 96/32478.
  • One may remove these sites by, for example, substituting or deleting residues, inserting residues into the site, or truncating portions containing the site.
  • the inserted or substituted residues may also be altered amino acids, such as peptidomimetics or D-amino acids.
  • Fc variants may be desirable for a number of reasons, several of which are described below.
  • Exemplary Fc variants include molecules and sequences in which:
  • cysteine-containing segment at the N-terminus may be truncated or cysteine residues may be deleted or substituted with other amino acids (e.g., alanyl, seryl).
  • cysteine residues may be deleted or substituted with other amino acids (e.g., alanyl, seryl).
  • one may truncate the N-terminal 20-amino acid segment of SEQ ID NO: 2 or delete or substitute the cysteine residues at positions 7 and 10 of SEQ ID NO: 2. Even when cysteine residues are removed, the single chain Fc domains can still form a dimeric Fc domain that is held together non-covalently.
  • a native Fc is modified to make it more compatible with a selected host cell. For example, one may remove the PA sequence near the N-terminus of a typical native Fc, which may be recognized by a digestive enzyme in E. coli such as proline iminopeptidase. One may also add an N-terminal methionine residue, especially when the molecule is expressed recombinantly in a bacterial cell such as E. coli.
  • the Fc domain of SEQ ID NO: 2 is one such Fc variant.
  • a portion of the N-terminus of a native Fc is removed to prevent N-terminal heterogeneity when expressed in a selected host cell. For this purpose, one may delete any of the first 20 amino acid residues at the N-terminus, particularly those at positions 1, 2, 3, 4 and 5.
  • Residues that are typically glycosylated may confer cytolytic response. Such residues may be deleted or substituted with unglycosylated residues (e.g., alanine).
  • a native Fc may have sites for interaction with certain white blood cells that are not required for the fusion molecules of the present invention and so may be removed.
  • ADCC site is removed.
  • ADCC sites are known in the art; see, for example, Molec. Immunol. 29 (5): 633-9 (1992) with regard to ADCC sites in IgG1. These sites, as well, are not required for the fusion molecules of the present invention and so may be removed.
  • the native Fc When the native Fc is derived from a non-human antibody, the native Fc may be humanized. Typically, to humanize a native Fc, one will substitute selected residues in the non-human native Fc with residues that are normally found in human native Fc. Techniques for antibody humanization are well known in the art.
  • Preferred Fc variants include the following.
  • the leucine at position 15 may be substituted with glutamate; the glutamate at position 99, with alanine; and the lysines at positions 101 and 103, with alanines.
  • one or more tyrosine residues can be replaced by phenyalanine residues.
  • An alternative vehicle would be a protein, polypeptide, peptide, antibody, antibody fragment, or small molecule (e.g., a peptidomimetic compound) capable of binding to a salvage receptor.
  • a polypeptide as described in U.S. Pat. No. 5,739,277, issued Apr. 14, 1998 to Presta et al.
  • Peptides could also be selected by phage display or RNA-peptide screening for binding to the FcRn salvage receptor.
  • salvage receptor-binding compounds are also included within the meaning of “vehicle” and are within the scope of this invention.
  • Such vehicles should be selected for increased half-life (e.g., by avoiding sequences recognized by proteases) and decreased immunogenicity (e.g., by favoring non-immunogenic sequences, as discovered in antibody humanization).
  • polymer vehicles may also be used for F 1 and F 2 .
  • Various means for attaching chemical moieties useful as vehicles are currently available, see, e.g., Patent Cooperation Treaty (“PCT”) International Publication No. WO 96/11953, entitled “N-Terminally Chemically Modified Protein Compositions and Methods,” herein incorporated by reference in its entirety.
  • PCT Patent Cooperation Treaty
  • This PCT publication discloses, among other things, the selective attachment of water soluble polymers to the N-terminus of proteins.
  • a preferred polymer vehicle is polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the PEG group may be of any convenient molecular weight and may be linear or branched.
  • the average molecular weight of the PEG will preferably range from about 2 kiloDalton (“kD”) to about 100 kD, more preferably from about 5 kD to about 50 kD, most preferably from about 5 kD to about 10 kD.
  • the PEG groups will generally be attached to the compounds of the invention via acylation or reductive alkylation through a reactive group on the PEG moiety (e.g., an aldehyde, amino, thiol, or ester group) to a reactive group on the inventive compound (e.g., an aldehyde, amino, or ester group).
  • a reactive group on the PEG moiety e.g., an aldehyde, amino, thiol, or ester group
  • a reactive group on the inventive compound e.g., an aldehyde, amino, or ester group
  • a useful strategy for the PEGylation of synthetic peptides consists of combining, through forming a conjugate linkage in solution, a peptide and a PEG moiety, each bearing a special functionality that is mutually reactive toward the other.
  • the peptides can be easily prepared with conventional solid phase synthesis.
  • the peptides are “preactivated” with an appropriate functional group at a specific site.
  • the precursors are purified and fully characterized prior to reacting with the PEG moiety.
  • Ligation of the peptide with PEG usually takes place in aqueous phase and can be easily monitored by reverse phase analytical HPLC.
  • the PEGylated peptides can be easily purified by preparative HPLC and characterized by analytical HPLC, amino acid analysis and laser desorption mass spectrometry.
  • Polysaccharide polymers are another type of water soluble polymer which may be used for protein modification.
  • Dextrans are polysaccharide polymers comprised of individual subunits of glucose predominantly linked by ⁇ 1-6 linkages. The dextran itself is available in many molecular 25 weight ranges, and is readily available in molecular weights from about 1 kD to about 70 kD.
  • Dextran is a suitable water soluble polymer for use in the present invention as a vehicle by itself or in combination with another vehicle (e.g., Fc). See, for example, WO 96/11953 and WO 96/05309. The use of dextran conjugated to therapeutic or diagnostic immunoglobulins has been reported; see, for example, European Patent Publication No. 0 315 456, which is hereby incorporated by reference in its entirety. Dextran of about 1 kD to about 20 kD is preferred when dextran is used as a vehicle in accordance with the present invention.
  • Linkers Any “linker” group is optional. When present, its chemical structure is not critical, since it serves primarily as a spacer.
  • the linker is preferably made up of amino acids linked together by peptide bonds.
  • the linker is made up of from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids. Some of these amino acids may be glycosylated, as is well understood by those in the art.
  • the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine.
  • a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine.
  • preferred linkers are polyglycines (particularly (Gly) 4 , (Gly) 5 ), poly(Gly-Ala), and polyalanines.
  • Other specific examples of linkers are: (Gly) 3 Lys(Gly) 4 ; (SEQ ID NO: 6) (Gly) 3 AsnGlySer(Gly) 2 ; (SEQ ID NO: 7) (Gly) 3 Cys(Gly) 4 ; and (SEQ ID NO: 8) GlyProAsnGlyGly. (SEQ ID NO: 9)
  • (Gly) 3 Lys(Gly) 4 means Gly-Gly-Gly-Lys-Gly-Gly-Gly-Gly. Combinations of Gly and Ala are also preferred.
  • the linkers shown here are exemplary; linkers within the scope of this invention may be much longer and may include other residues.
  • Non-peptide linkers are also possible.
  • These alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., C 1 -C 6 ) lower acyl, halogen (e.g., Cl, Br), CN, NH 2 , phenyl, etc.
  • An exemplary non-peptide linker is a PEG linker, VI
  • n is such that the linker has a molecular weight of 100 to 5000 kD, preferably 100 to 500 kD.
  • the peptide linkers may be altered to form derivatives in the same manner as described above.
  • the compound or some portion thereof is cyclic.
  • the peptide portion may be modified to contain two or more Cys residues (e.g., in the linker), which could cyclize by disulfide bond formation.
  • the compound is cross-linked or is rendered capable of cross-linking between molecules.
  • the peptide portion may be modified 20 to contain one Cys residue and thereby be able to form an intermolecular disulfide bond with a like molecule.
  • the compound may also be cross-linked through its C-terminus, as in the molecule shown below.
  • One or more peptidyl [—C(O)NR—] linkages (bonds) is replaced by a non-peptidyl linkage.
  • Exemplary non-peptidyl linkages are —CH 2 -carbamate [—CH 2 —OC(O)NR—], phosphonate, —CH 2 -sulfonamide [—CH 2 —S(O) 2 NR—], urea [—NHC(O)NH—], —CH 2 -secondary amine, and alkylated peptide [—C(O)NR 6 — wherein R 6 is lower alkyl].
  • the N-terminus is derivatized. Typically, the N-terminus may be acylated or modified to a substituted amine.
  • Exemplary N-terminal derivative groups include —NRR 1 (other than —NH 2 ), —NRC(O)R 1 , —NRC(O)OR 1 , —NRS(O) 2 R 1 , —NHC(O)NHR 1 , succinimide, or benzyloxycarbonyl-NH— (CBZ-NH—), wherein R and R 1 are each independently hydrogen or lower alkyl and wherein the phenyl ring may be substituted with 1 to 3 substituents selected from the group consisting of C 1 -C 4 alkyl, C 1 -C 4 alkoxy, chloro, and bromo.
  • the free C-terminus is derivatized. Typically, the C-terminus is esterified or amidated.
  • Exemplary C-terminal derivative groups include, for example, —C(O)R 2 wherein R 2 is lower alkoxy or —NR 3 R 4 wherein R 3 and R 4 are independently hydrogen or C 1 -C 8 alkyl (preferably C 1 -C 4 alkyl).
  • a disulfide bond is replaced with another, preferably more stable, cross-linking moiety (e.g., an alkylene). See, e.g., Bhatnagar et al. (1996), J. Med. Chem. 39: 3814-9; Alberts et al. (1993) Thirteenth Am. Pep. Symp., 357-9.
  • another, preferably more stable, cross-linking moiety e.g., an alkylene
  • One or more individual amino acid residues is modified.
  • Various derivatizing agents are known to react specifically with selected sidechains or terminal residues, as described in detail below.
  • Lysinyl residues and amino terminal residues may be reacted with succinic or other carboxylic acid anhydrides, which reverse the charge of the lysinyl residues.
  • suitable reagents for derivatizing alpha-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate.
  • Arginyl residues may be modified by reaction with any one or combination of several conventional reagents, including phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginyl residues requires that the reaction be performed in alkaline conditions because of the high pKa of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group.
  • Carboxyl sidechain groups may be selectively modified by reaction with carbodiimides (R′—N ⁇ C ⁇ N—R′) such as 1-cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl) carbodiimide.
  • carbodiimides R′—N ⁇ C ⁇ N—R′
  • aspartyl and glutamyl residues may be converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
  • Glutaminyl and asparaginyl residues may be deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
  • Cysteinyl residues can be replaced by amino acid residues or other moieties either to eliminate disulfide bonding or, conversely, to stabilize cross-linking. See, e.g., Bhatnagar et al. (1996), J. Med. Chem. 39: 3814-9.
  • Derivatization with bifunctional agents is useful for cross-linking the peptides or their functional derivatives to a water-insoluble support matrix or to other macromolecular vehicles.
  • Commonly used cross-linking agents include, e.g., 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3′-dithiobis(succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane.
  • Derivatizing agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light.
  • reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization.
  • Carbohydrate (oligosaccharide) groups may conveniently be attached to sites that are known to be glycosylation sites in proteins.
  • O-linked oligosaccharides are attached to serine (Ser) or threonine (Thr) residues while N-linked oligosaccharides are attached to asparagine (Asn) residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline.
  • X is preferably one of the 19 naturally occurring amino acids other than proline.
  • the structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different.
  • sialic acid is usually the terminal residue of both N-linked and O-linked oligosaccharides and, by virtue of its negative charge, may confer acidic properties to the glycosylated compound.
  • site(s) may be incorporated in the linker of the compounds of this invention and are preferably glycosylated by a cell during recombinant production of the polypeptide compounds (e.g., in mammalian cells such as CHO, BHK, COS). However, such sites may further be glycosylated by synthetic or semi-synthetic procedures known in the art.
  • Compounds of the present invention may be changed at the DNA level, as well.
  • the DNA sequence of any portion of the compound may be changed to codons more compatible with the chosen host cell.
  • optimized codons are known in the art. Codons may be substituted to eliminate restriction sites or to include silent restriction sites, which may aid in processing of the DNA in the selected host cell.
  • the vehicle, linker and peptide DNA sequences may be modified to include any of the foregoing sequence changes.
  • the compounds of this invention largely may be made in transformed host cells using recombinant DNA techniques.
  • a recombinant DNA molecule coding for the peptide is prepared.
  • Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidate method. Also, a combination of these techniques could be used.
  • the invention also includes a vector capable of expressing the peptides in an appropriate host.
  • the vector comprises the DNA molecule that codes for the peptides operatively linked to appropriate expression control sequences. Methods of effecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known.
  • Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation.
  • the resulting vector having the DNA molecule thereon is used to transform an appropriate host. This transformation may be performed using methods well known in the art.
  • Any of a large number of available and well-known host cells may be used in the practice of this invention.
  • the selection of a particular host is dependent upon a number of factors recognized by the art. These include, for example, compatibility with the chosen expression vector, toxicity of the peptides encoded by the DNA molecule, rate of transformation, ease of recovery of the peptides, expression characteristics, bio-safety and costs. A balance of these factors must be struck with the understanding that not all hosts may be equally effective for the expression of a particular DNA sequence.
  • useful microbial hosts include bacteria (such as E. coli sp.), yeast (such as Saccharomyces sp.) and other fungi, insects, plants, mammalian (including human) cells in culture, or other hosts known in the art.
  • the transformed host is cultured and purified.
  • Host cells may be cultured under conventional fermentation conditions so that the desired compounds are expressed. Such fermentation conditions are well known in the art.
  • the peptides are purified from culture by methods well known in the art.
  • the compounds may also be made by synthetic methods.
  • solid phase synthesis techniques may be used. Suitable techniques are well known in the art, and include those described in Merrifield (1973), Chem. Polypeptides, pp. 335-61 (Katsoyannis and Panayotis eds.); Merrifield (1963), J. Am. Chem. Soc. 85: 2149; Davis et al. (1985), Biochem. Intl. 10: 394-414; Stewart and Young (1969), Solid Phase Peptide Synthesis; U.S. Pat. No. 3,941,763; Finn et al. (1976), The Proteins (3rd ed.) 2: 105-253; and Erickson et al. (1976), The Proteins (3rd ed.) 2: 257-527. Solid phase synthesis is the preferred technique of making individual peptides since it is the most cost-effective method of making small peptides.
  • the compounds of this invention have pharmacologic activity resulting from their interaction with PTH-1 receptor or PTH-2 receptor. Mannstadt et al. (1999), Am. J. Physiol. 277. 5Pt 2. F665-75. PTH and agonists thereof increase bone resorption, increase renal calcium reabsorption, decrease epidermal proliferation, and decrease hair growth. Holick et al. (1994) Proc. Natl. Sci. USA 91 (17): 8014-6; Schilli et al. (1997), J. Invest. Dermatol. 108(6): 928-32. Thus, antagonists of PTH-1 receptor and/or PTH-2 receptor are useful in treating:
  • hypercalcemia including hypercalcemia resulting from solid tumors (breast, lung and kidney) and hematologic malignacies (multiple myeloma, lymphoma and leukemia); idiopathic hypercalcemia, and hypercalcemia associated with hyperthyroidism and renal function disorders;
  • tumor metastases particularly metastases to bone, and particularly related to breast and prostate cancer
  • osteopenia that is related to or aggravated by aberrant PTH receptor signaling, including various forms of osteoporosis, such as:
  • osteoporosis e.g., osteogenesis imperfecta, homocystinuria, Menkes' syndrome, and Riley-Day syndrome
  • osteoporosis secondary to other disorders such as hemochromatosis, hyperprolactinemia, anorexia nervosa, thyrotoxicosis, diabetes mellitus, celiac disease, inflammatory bowel disease, primary biliary cirrhosis, rheumatoid arthritis, ankylosing spondylitis, multiple myeloma, lymphoproliferative diseases, and systemic mastocytosis;
  • osteoporosis secondary to surgery e.g., gastrectomy
  • drug therapy such as chemotherapy, anticonvulsant therapy, immunosuppressive therapy, and anticoagulant therapy
  • osteoporosis secondary to glucocorticosteroid treatment for such diseases as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), asthma, temporal arteritis, vasculitis, chronic obstructive pulmonary disease, polymyalgia rheumatica, polymyositis, chronic interstitial lung disease;
  • RA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • asthma systemic lupus erythematosus
  • vasculitis chronic obstructive pulmonary disease
  • polymyalgia rheumatica polymyositis
  • chronic interstitial lung disease for such diseases as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), asthma, temporal arteritis, vasculitis, chronic obstructive pulmonary disease, polymyalgia r
  • osteoporosis secondary to glucocorticosteroid and/or immunomodulatory treatment to prevent organ rejection following organ transplant such as kidney, liver, lung, heart transplants;
  • osteoporosis associated with malignant disease such as breast cancer, prostate cancer
  • osteomyelitis or an infectious lesion in bone, leading to bone loss
  • osteopenia following surgery, induced by steroid administration, and associated with disorders of the small and large intestine and with chronic hepatic and renal diseases.
  • alopecia deficient hair growth or partial or complete hair loss
  • alopecia including androgenic alopecia (male pattern baldness), toxic alopecia, alopecia senilis, alopecia areata, alopecia pelada, and trichotillomania
  • PTH receptor agonists are useful as a therapeutic treatment.
  • such indications include fracture repair (including healing of non-union fractures), osteopenia, including various forms of osteoporosis, such as:
  • osteoporosis e.g., osteogenesis imperfecta, homocystinuria, Menkes' syndrome, and Riley-Day syndrome
  • osteoporosis secondary to other disorders such as hemochromatosis, hyperprolactinemia, anorexia nervosa, thyrotoxicosis, diabetes mellitus, celiac disease, inflammatory bowel disease, primary biliary cirrhosis, rheumatoid arthritis, ankylosing spondylitis, multiple myeloma, lymphoproliferative diseases, and systemic mastocytosis;
  • osteoporosis secondary to surgery e.g., gastrectomy
  • drug therapy such as chemotherapy, anticonvulsant therapy, immunosuppressive therapy, and anticoagulant therapy
  • osteoporosis secondary to glucocorticosteroid treatment for diseases such as RA, SLE, asthma, temporal arteritis, vasculitis, chronic obstructive pulmonary disease, polymyalgia rheumatica, polymyositis, chronic interstitial lung disease;
  • osteoporosis secondary to glucocorticosteroid and/or immunomodulatory treatment to prevent organ rejection following organ transplant such as kidney, liver, lung, heart transplants;
  • osteoporosis associated with malignant disease such as breast cancer, prostate cancer
  • PTH agonists with extended half-life may be used with an inhibitor of bone resorption.
  • Inhibitors of bone resorption include OPG and OPG derivatives, OPG-L (RANKL) antibody, calcitonin (e.g., Miacalcin®, Calcimar®), bisphosphonates (e.g., APD, alendronate, risedronate, etidronate, pamidronate, tiludronate, clodronate, neridronate, ibandronate, zoledronate), estrogens (e.g., Premarin®, Estraderm®, Prempro®, Alora®, Climara®, Vivelle®, Estratab® Ogen®), selective estrogen receptor modulators (e.g., raloxifene, droloxifene, lasofoxifene), tibolone, and the like.
  • Exemplary bone resorption inhibitors are
  • the compounds of this invention may be appropriate as a monotherapy for the treatment of Osteoporosis, and it is possible that the addition of an antiresorptive agent to PTH-Fc treatment will increase both their efficacy and therapeutic window. Both PTH and PTH-Fc cause an increase in both bone formation and bone resorption. The ability of antiresorptives to block the osteoclast response could limit the hypercalcemic effects of PTH-Fc and could also increase bone mas
  • the present invention also provides methods of using pharmaceutical compositions of the inventive compounds.
  • Such pharmaceutical compositions may be for administration for injection, or for oral, pulmonary, nasal, transdermal or other forms of administration.
  • the invention encompasses pharmaceutical compositions comprising effective amounts of a compound of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers.
  • compositions include diluents of various buffer content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc. or into liposomes.
  • buffer content e.g., Tris-HCl, acetate, phosphate
  • additives e.g., Tween 80, Polysorbate 80
  • anti-oxidants e.g., ascorbic acid, sodium metabisulfite
  • preservatives e.g., Thimersol, benzyl alcohol
  • Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation.
  • 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 in their entirety.
  • the compositions may be prepared in liquid form, or may be in dried powder, such as lyophilized form. Implantable sustained release formulations are also contemplated, as are transdermal formulations.
  • Twice weekly dosing of the compounds of this invention is superior to daily injection of PTH (1-34) for increasing osteoblast number, bone volume, and bone mineral density in rodents.
  • PTH-(1-34)-Fc caused greater increases in bone density and bone volume compared to daily PTH-(1-34).
  • FIG. 10. In an aged OVX rat model of osteoporosis, twice weekly dosing was able to reverse more than 50% of the bone loss induced by one year of estrogen ablation. The effect seen in the aged rat model was even greater when combined with a bisphosphonate (APD).
  • the optimal dosing of primates may be less frequent compared to rats or mice. Weekly (or less frequent) dosing may be optimal in primates, based on the observation that the hypercalcemic response of OVX cynomolgus monkeys to a single subcutaneous injection of PTH-(1-34)-Fc (10-34 nmol/kg) persisted for about 168 hours (FIG. 11). This observation suggests that a single subcutaneous dose of PTH-(1-34)-Fc in primates is cleared within about 1 week, which could also represent the maximum dosing frequency required for anabolic effects.
  • Oral dosage forms Contemplated for use herein are oral solid dosage forms, which are described generally in Chapter 89 of Remington's Pharmaceutical Sciences (1990), 18th Ed., Mack Publishing Co. Easton Pa. 18042, which is herein incorporated by reference in its entirety. Solid dosage forms include tablets, capsules, pills, troches or lozenges, cachets or pellets. Also, liposomal or proteinoid encapsulation may be used to formulate the present compositions (as, for example, proteinoid microspheres reported in U.S. Pat. No. 4,925,673). Liposomal encapsulation may be used and the liposomes may be derivatized with various polymers (e.g., U.S. Pat. No.
  • the formulation will include the inventive compound, and inert ingredients which allow for protection against the stomach environment, and release of the biologically active material in the intestine.
  • the compounds may be chemically modified so that oral delivery is efficacious.
  • the chemical modification contemplated is the attachment of at least one moiety to the compound molecule itself, where said moiety permits (a) inhibition of proteolysis; and (b) uptake into the blood stream from the stomach or intestine.
  • the increase in overall stability of the compound and increase in circulation time in the body are also be used for this purpose.
  • moieties include: PEG, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. See, for example, Abuchowski and Davis, Soluble Polymer - Enzyme Adducts Enzymes as Drugs (1981), Hocenberg and Roberts, eds., Wiley-Interscience, New York, N.Y., pp. 367-83; Newmark, et al. (1982), J. Appl. Biochem. 4:185-9.
  • Other polymers that could be used are poly-1,3-dioxolane and poly-1,3,6-tioxocane.
  • Preferred for pharmaceutical usage, as indicated above, are PEG moieties.
  • a salt of a modified aliphatic amino acid such as sodium N-(8-[2-hydroxybenzoyl]amino) caprylate (SNAC)
  • SNAC sodium N-(8-[2-hydroxybenzoyl]amino) caprylate
  • the compounds of this invention can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about 1 mm.
  • the formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets.
  • the therapeutic could be prepared by compression.
  • Colorants and flavoring agents may all be included.
  • the protein (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 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 disintegrants include but are not limited to starch including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used.
  • Another form of the disintegrants are the insoluble cationic exchange resins.
  • Powdered gums may be used as disintegrants and as binders and these 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 therapeutic 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 hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.
  • MC methyl cellulose
  • EC ethyl cellulose
  • CMC carboxymethyl cellulose
  • PVP polyvinyl pyrrolidone
  • HPMC hydroxypropylmethyl cellulose
  • An antifrictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process.
  • Lubricants may be used as a layer between the therapeutic 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.
  • Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added.
  • the glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.
  • 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.
  • anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
  • Cationic detergents might be used and could include benzalkonium chloride or benzethonium 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 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 may also be included in the formulation to enhance uptake of the compound. Additives potentially having this property are for instance the fatty acids oleic acid, linoleic acid and linolenic acid.
  • Controlled release formulation may be desirable.
  • the compound of this invention 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, e.g., alginates, polysaccharides.
  • Another form of a controlled release of the compounds of this invention 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. Some enteric coatings also have a delayed release effect.
  • coatings may be used for the formulation. These include a variety of sugars which could be applied in a coating pan.
  • the therapeutic agent 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 nonenteric 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 phthalic 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.
  • Pulmonary delivery forms are also contemplated herein.
  • the protein (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream.
  • Adjei et al. Pharma. Res. (1990) 7: 565-9
  • Adjei et al. (1990), Internatl. J. Pharmaceutics 63: 135-44 (leuprolide acetate); Braquet et al. (1989), J. Cardiovasc. Pharmacol. 13 (suppl.5): s.143-146 (endothelin-1); Hubbard 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, manufactured by Mallinckrodt, Inc., St. Louis, Mo.; the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colo.; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, N.C.; and the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Mass.
  • each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to diluents, adjuvants and/or carriers useful in therapy.
  • inventive compound should most advantageously be prepared in particulate form with an average particle size of less than 10 ⁇ m (or microns), most preferably 0.5 to 5 ⁇ m, for most effective delivery to the distal lung.
  • Pharmaceutically acceptable carriers include carbohydrates such as trehalose, mannitol, xylitol, sucrose, lactose, and sorbitol.
  • Other ingredients for use in formulations may include DPPC, DOPE, DSPC and DOPC.
  • Natural or synthetic surfactants may be used.
  • PEG may be used (even apart from its use in derivatizing the protein or analog).
  • Dextrans such as cyclodextran, may be used.
  • Bile salts and other related enhancers may be used.
  • Cellulose and cellulose derivatives may be used. Amino acids may be used, such as use in a buffer formulation.
  • liposomes are contemplated.
  • Formulations suitable for use with a nebulizer will typically comprise the inventive compound 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 protein 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 inventive compound 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 trichlorofluoromethane, 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 the inventive compound and may also include a bulking agent, such as lactose, sorbitol, sucrose, mannitol, trehalose, or xylitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.
  • a bulking agent such as lactose, sorbitol, sucrose, mannitol, trehalose, or xylitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.
  • Nasal delivery forms Nasal delivery of the inventive compound is also contemplated. Nasal delivery allows the passage of the protein 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. Delivery via transport across other mucous membranes is also contemplated.
  • the dosage regimen involved in a method for treating the above-described conditions will be determined by the attending physician, considering various factors which modify the action of drugs, e.g. the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors.
  • the daily regimen should be in the range of 0.1-1000 micrograms of the inventive compound per kilogram of body weight, preferably 0.1-150 micrograms per kilogram.
  • the inventors have determined preferred structures for the preferred peptides listed in Table 4 below.
  • the symbol “A” may be any of the linkers described herein or may simply represent a normal peptide bond (i.e., so that no linker is present). Tandem repeats and linkers are shown separated by dashes for clarity.
  • F 1 is an Fc domain as defined previously herein.
  • the inventors further contemplate heterodimers in which each strand of an Fc dimer is linked to a different peptide sequence; for example, a molecule in which one strand can be described by SEQ ID NO: 166, the other by SEQ ID NO: 170 or an Fc linked with any of the sequences in Tables 1 and 2.
  • Parathyroid hormone [PTH-(1-34) or native PTH-(1-84)] causes increased bone formation and increased bone mass when injected daily. This anabolic response was previously thought to require brief exposure to PTH, which is facilitated by the short half-life (less than 1 h) of PTH. Clinically, the anabolic effect of PTH therapy requires daily SC injection, which is a significant barrier to the widespread use of PTH. Less frequent injections of PTH would be clinically desirable and could be achieved by increasing the in vivo half-life of PTH.
  • Co-administration of a potent bone resorption inhibitor may provide greater effect.
  • This regimen would theoretically permit the unopposed stimulation of bone formation by PTH, leading to increased bone mass.
  • other bone resorption inhibitors including bisphosphonates or estrogen, would also inhibit PTH-induced bone resorption and could therefore be used in combination with a long-acting PTH molecule.
  • PTH-(1-34)-Fc Fc conjugation of proteins causes a significant increase in their circulating half life, which may permit injections of PTH-(1-34)-Fc on a schedule similar to or identical to that of OPG-Fc.
  • the benefits of this invention include less frequent injections of PTH, from the current standard of once per day to as infrequently as once per quarter.
  • Hypercalcemia induced by PTH-(1-34) persisted for 6-24 h, while equimolar doses of PTH-(1-34)-Fc caused more sustained hypercalcemia (48-72 h).
  • Group 1 Vehicle controls (PBS, injected SC, Days 1, 3, and 5)
  • Group 2 OPG-Fc, single SC injection (1 mg/kg) on Day 1
  • Group 3 PTH-(1-34), SC injections on Days 1, 3, and 5, at 20 nMoles PTH/kg/injection. This represents an optimal anabolic PTH regimen.
  • Group 4 Same as group 3, but with a single OPG-Fc injection on Day 1.
  • Group 5 Single SC injection of PTH-(1-34)-Fc at 60 nMoles/kg, on Day 1. This represents a molar dose which is equivalent to the total dose of PTH-(1-34) received by group 3, but in a single injection.
  • Group 6 Same as group 5, but with a single OPG-Fc injection (SC, 1 mg/kg) on Day 1.
  • BMD in L3 increased modestly with a single injection of OPG-Fc, or with 3 injections of PTH-(1-34), compared to PBS-treated rats (FIG. 5).
  • PTH-(1-34)+OPG caused a greater increase in BMD than either OPG or PTH-(1-34) alone.
  • a single injection of PTH-(1-34)-Fc failed to increase BMD.
  • a single injection of PTH-(1-34)-Fc plus a single injection of OPG-Fc caused a significant increase in BMD (FIG. 5).
  • FIG. 5 shows the effect of PTH-Fc +OPG-Fc on bone mineral density (BMD) in the third lumbar vertebra (L3).
  • BMD bone mineral density
  • PTH-(7-34) peptides are fairly effective PTH-R1 antagonists with very mild agonist activity. Compared to PTH-(7-34), PTHrP-(7-34) peptide has greater affinity for PTH-R1 and as such is a more potent antagonist. However, PTHrP-(7-34) also has greater (but still mild) agonist activity compared to PTH-(7-34) (McKee (1990), Endocrinol. 127: 76). The optimal antagonist may combine the weaker agonism of PTH-(7-34) with the stronger antagonism of PTHrP-(7-34).
  • mice were then treated once SC with varying doses of [Asn10,Leu11]PTHrP-(7-34)-Fc, or with vehicle (PBS).
  • vehicle-treated mice challenged with PTHrP-(1-34) a transient hypercalcemic response was observed. The peak calcemic response occurred at 3 h post challenge, and persisted until at least 6 h post challenge.
  • [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 mg/kg caused a more rapid normalization of PTHrP-induced hypercalcemia compared to vehicle treatment. A dose of 30 mg/kg completely blocked the calcemic response to PTHrP-(1-34) (FIG. 6).
  • mice [0326] We then tested the effects of PTH-(1-34), PTH-(1-34)-Fc, PTH-(1-31)-Fc and PTH-(1-30)-Fc in mice.
  • Four week old male mice were injected on days 0, 5, and 10 with vehicle or with PTH fragments, by SC injection.
  • Peripheral blood was obtained for clinical chemistry at 24, 48, and 72 h.
  • Mice were killed on day 15, vertebrae, tibiae and femurs were harvested for histology and one tibia was collected for bone density measurements (peripheral quantitative computed tomography, pQCT).
  • Clinical chemistry endpoints included total serum calcium, serum alkaline phosphatase (AP, a marker of osteoblast activity), and serum tartrate-resistant acid phosphatase (TRAP, a marker of osteoclast activity). For each animal, the ratio of AP:TRAP was calculated as an index of relative osteoblast activity compared to osteoclast activity. A higher AP:TRAP ratio would indicate a potentially more anabolic agent.
  • the relatively high doses (15-fold greater than optimal anabolic doses) were selected base on previous studies which demonstrated significant changes in clinical chemistry endpoints. It was anticipated that lower doses might be required to demonstrate anabolic effects on bone density, and that antiresorptive co-treatment might also be required to achieve anabolic responses.
  • Serum AP (osteoblast marker) was unchanged by PTH-(1-34) administration, but was significantly elevated by 300 nmoles/kg of PTH-(1-34)-Fc and by PTH-(1-31)-Fc at 72 h. PTH-(1-30)-Fc demonstrated the greatest elevation of AP, which peaked 72 h after injection of 1,000 nmoles/kg (FIG. 8B). Serum TRAP (osteoclast marker) was not significantly changed by PTH-(1-34), PTH-(1-34)-Fc, or PTH-(1-30)-Fc, but was dramatically increased by PTH-(1-31)-Fc (FIG. 8C).
  • the calculated AP:TRAP ratios were unchanged by PTH-(1-34), and were increased over time by PTH-(1-34)-Fc.
  • the low dose of PTH-(1-31)-Fc (100 nmoles/kg) increased AP:TRAP, while the high dose (1,000 nmoles/kg) decreased AP:TRAP.
  • the greatest increase in AP:TRAP was realized with PTH-(1-30)-Fc (1,000 nmoles/kg) (FIG. 8D).
  • PTH-(1-30)-Fc caused the greatest increase in bone density.
  • the reverse dose-response was consistent with the notion that doses employed (chosen for clinical chemistry endpoints) were 5-50 fold higher than the optimal anabolic doses.
  • Low doses of PTH (or PTH-Fc) which fail to significantly increase serum calcium are optimal for anabolic effects. See Hock, J. M. (1992), J. Bone Min. Res. 7:65-72.
  • the treatment regimen with the greatest anabolic effect was also the only PTH-Fc treatment which failed to significantly increase serum calcium (FIG. 8A).
  • mice were treated twice per week by subcutaneous injection with various doses of PTH-(1-34)-Fc or with vehicle (PBS). Other mice were treated daily with SC injections of PTH-(1-34) at a dose of 80 ⁇ g/kg/day (20 nmol/kg/day), a treatment regimen which is optimal for increasing bone mass in rodents (M. Gunness-Hey and J. M. Hock, Metab. Bone Dis. & Rel. Res. 5:177-181, 1984). After 3 weeks, mice were sacrificed and tibiae were analyzed for bone mineral density (BMD) via pQCT (FIG. 10).
  • BMD bone mineral density
  • Twice-weekly PTH-(1-34)-Fc also effectively increased BMD as a monotherapy in aged ovariectomized (OVX) rats.
  • OVX ovariectomized rats
  • Sprague Dawley rats were OVX'd at 3 months of age and allowed to lose bone for 11 months.
  • Other rats were sham-operated and treated twice per week with vehicle (PBS).
  • OVX rats were treated twice per week with SC injections of either vehicle or the bisphosphonate APD (pamidronate, 0.5 mg/kg), or with PTH-(1-34)-Fc (50 nmol/kg) or with APD+PTH-(1-34)-Fc.
  • BMD was determined weekly via dual energy X-ray absorptiometry (DEXA).
  • Rats were sacrificed after 4 weeks of treatment.
  • OVX rats had significant reductions in BMD at all skeletal sites analyzed, compared to vehicle-treated OVX rats (FIG. 11, p ⁇ 0.05, 2-way ANOVA).
  • APD alone did not significantly increase BMD at any skeletal site compared to vehicle-treated OVX rats.
  • PTH-(1-34)-Fc alone caused a significant increase in BMD at the femoral metaphysis after 4 weeks of treatment (p ⁇ 0.05).
  • Treatment of OVX rats with PTH+ABD was associated with an earlier significant increase in BMD at this site (3 weeks).
  • PTH-(1-34)-Fc is an effective anabolic agent when used as a monotherapy in both adult mice and aged OVX rats.
  • APD antiresorptive agent
  • Co-administration of APD also blocked the transient hypercalcemic response produced by PTH-(1-34)-Fc, which suggests that the therapeutic index of PTH-(1-34)-Fc could be significantly improved by co-administering an effective antiresorptive agent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Molecular Biology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Pulmonology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Obesity (AREA)
  • Oncology (AREA)
  • Cardiology (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)

Abstract

The present invention concerns therapeutic agents that modulate the activity of PTH and PTHrP. In accordance with the present invention, modulators of PTH and PTHrP comprise:
(a) a PTH/PTHrP modulating domain; and
(b) a vehicle, such as a polymer (e.g., PEG or dextran) or an Fc domain, which is preferred;
wherein the vehicle is covalently attached to the C-terminus of the PTH/PTHrP modulating domain. The vehicle and the PTH/PTHrP modulating domain may be linked through the N— or C-terminus of the PTH/PTHrP modulating domain, as described further below. The preferred vehicle is an Fc domain, and the preferred Fc domain is an IgG Fc domain. Preferred PTH/PTHrP modulating domains comprise the PTH and PTHrP-derived amino acid sequences described hereinafter. Other PTH/PTHrP modulating domains can be generated by phage display, RNA-peptide screening and the other techniques mentioned herein. Such peptides typically will be modulators of both PTH activity and PTHrP activity, although such techniques can be used to generate peptide sequences that serve as selective modulators (e.g., agonists of PTH activity but not PTHrP activity).

Description

  • This application is a divisional of U.S. application Ser. No. 09/843,221, filed Apr. 26, 2001, which claims the benefit of U.S. Provisional Application No. 60/266,673, filed Feb. 6, 2001, U.S. Provisional Application No. 60/214,860, filed Jun. 28, 2000, and U.S. Provisional Application No. 60/200,053, filed Apr. 27, 2000, which are hereby incorporated by reference.[0001]
  • BACKGROUND OF THE INVENTION
  • Parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP) play important physiological roles in calcium homeostasis and in development, respectively. Calcium concentration in the blood is tightly regulated, due to the essential role of calcium in cell metabolism. PTH is an endocrine hormone which is secreted from the parathyroid gland in response to decreased serum calcium levels. PTH acts directly to increase bone resorption and to stimulate renal calcium reabsorption, thus increasing or preserving circulating calcium stores. PTH also indirectly increases calcium absorption in the gut by stimulating the renal hydroxylation of vitamin D. [0002]
  • Both primary and secondary hyperparathyroidism are conditions that are associated with excessive levels of circulating parathyroid hormone. Through the aforementioned pathways, excess PTH levels can cause hypercalcemia and osteopenia. Bone resorption inhibitors such as bisphosphonates and OPG can effectively protect bone and can inhibit the skeleton's contribution to hypercalcemia. However, the calcemic effects of hyperparathyroidism on the kidney and gut are not addressed by currently available therapy. [0003]
  • PTHrP is produced by many cell types, and plays an important role in regulating skeletal development. Postnatally, the roles for PTHrP are less clearly defined. Circulating levels of PTHrP are essentially non-detectable in normal healthy adults. However, many tumors of diverse embryological origins produce and secrete PTHrP in quantities sufficient to cause hypercalcemia. In fact, humoral hypercalcemia of malignancy (HHM) is the most common paraneoplastic syndrome, which accounts for significant patient morbidity and mortality. [0004]
  • Currently, HHM is treated with saline hydration followed by bone resorption inhibitors such as bisphosphonates. This treatment regimen typically takes 3-4 days to achieve significant reductions in serum calcium, and the effects are relatively short-lived (less than one month). For patients with high circulating levels of PTHrP, the effects of current treatment options are even less impressive. Repeated administration of conventional therapies are usually progressively less effective. These limitations to current therapy strongly indicate an unmet medical need for rapid, effective, and long-lasting treatments for HHM. [0005]
  • A major reason for the limited benefits of current HHM therapy is the failure to directly inhibit PTHrP, which is very well established as the principal pathophysiologic factor in HHM. Bone resorption inhibitors such as bisphosphonates only inhibit bone resorption, while PTHrP also has significant calcemic effects on the kidney and the gut. Total neutralization of PTHrP would be the ideal adjuvant therapeutic approach to treatment of HHM. [0006]
  • Both PTH and PTHrP interact with PTH-1 receptor, which accounts for most of their known effects. Mannstadt et al. (1999), [0007] Am. J. Physiol. 277. 5Pt 2. F665-75 (1999). Only PTH interacts with the newly discovered PTH-2 receptor. Id. PTHrP can be changed to a PTH-2 receptor agonist, however, by changing two residues to the residues at those positions in PTH. Gardella et al. (1996), J. Biol. Chem. 271 (33): 19888-93.
  • An N-terminal fragment of PTH has been used as a therapeutic agent. Intermittently administered native PTH-(1-84) exhibits osteogenic properties, and it has been recognized for decades that these properties can be fully realized with the C-terminally truncated fragment PTH-(1-34). Both peptides bind and activate the PTH-1 receptor with similar affinities, causing the activation of adenylate cyclase (AC) as well as phospholipase C (PLC). AC activation through PTH-1 receptor generates cAMP, while PLC activation through PTH-1 receptor generates PKC and intracellular calcium transients. PTH-(1-34) can maximally activate both the AC and the PLC pathways. It has been demonstrated that the anabolic effects of PTH-(1-34) require short intermittent (daily) exposures Dobnig (1998), [0008] Endocrinol. 138: 4607-12. In human trials on postmenopausal women, daily subcutaneous injection of low doses of PTH(1-34) were shown to result in impressive bone formation in the spine and femoral neck with significant reduction in incidence of vertebral fractures. These clinical data reveal PTH as one of the most efficacious agents tested for osteoporosis.
  • Truncated PTH fragments have diminished AC/cAMP activation and similarly diminished anabolic activity. Rixon et al. (1994), [0009] J. Bone Min. Res. 9: 1179-89; Hilliker et al. (1996), Bone 19: 469-477; Lane et al. (1996), J. Bone Min. Res. 11: 614-25. Such truncated PTH fragments have this diminished activity (Rixon et al. (1994); Hilliker et al. (1996); Lane et al. (1996)) even if they maintain full agonism towards PKC. Rixon et al., (1994). These observations have led to the proposal that the AC/cAMP pathway is critical for the bone anabolic properties of PTH, while the PLC/PKC pathway is dispensable in this regard. Rixon et al., (1994); Whitfield et al. (1996), Calcified Tissue International 53: 81-7.
  • An opposing, but not mutually exclusive, theory suggests that PLC activation (in addition to AC) might also be an important property of anabolic PTH fragments. Takasu (1998), [0010] Endocrinol. 139: 4293-9. The apparent absence of PLC activation by some anabolic C-terminally truncated PTH peptides may be an artifact of insensitive assay methods combined with lower receptor binding. Takasu (1998). Progressive truncations from the C-terminus of PTH-(1-34) result in stepwise reductions in binding affinity for the PTH1R Takasu (1998). PKC activation through PTH-1 receptor appears to be acutely sensitive to binding affinity and to receptor density (Guo et al. (1995), Endocrinol 136: 3884-91), whereas cAMP activation is far less sensitive to these variables. As such, hPTH-(1-31) has a slightly reduced (1-6 fold) affinity for PTH-1 receptor compared to hPTH-(1-34), while hPTH-(1-30) has a significantly reduced (10-100 fold) affinity Takasu (1998). Perhaps due to this decreased PTH-1 receptor affinity, PTH-(1-30) is a weak and incomplete agonist for PLC activation via the rat PTH-1 receptor.
  • Compared to PTH-(1-34), PTH-(1-31) has similar or slightly reduced anabolic potential (Rixon et al. (1994); Whitfield et al. (1996), [0011] Calcified Tissue International 53: 81-7; Whitfield et al. (1996), Calcified Tissue International 65: 143-7), binding affinity for PTH1R, and cAMP induction (Takasu (1998)). PTH-(1-31) also has slightly reduced PLC activation. Takasu (1998). In healthy humans, infusion of PTH-(1-31) and PTH-(1-34) had similar stimulatory effects on plasma and urinary cAMP concentration, but unlike PTH-(1-34), PTH-(1-31) failed to elevate serum calcium, plasma 1,25(OH)2D3, or urinary N-TX levels. Fraher et al. (1999), J. Clin. Endocrin. Met. 84: 2739-43. These data suggest that PTH-(1-31) has diminished capacity to induce bone resorption and to stimulation vitamin D synthesis, which is a favorable profile for bone anabolic agents.
  • PTH-(1-30) was initially shown to lack anabolic properties Whitfield et al. (1996), [0012] Calcified Tissue International 53: 81-7. More recently, however, it has been demonstrated that PTH-(1-30) is anabolic when administered at very high doses (400-2,000 μg/kg, vs. 80 μg/kg for PTH-(1-34)). The lower potency of PTH-(1-30) could be predicted by its lower binding affinity for PTH-1 receptor, its diminished cAMP activation, and/or to its greatly diminished PKC activation. Takasu (1998). It remains to be determined whether PTH-(1-30) has a similar or even more desirable reduction in apparent bone resorption activity.
  • PTH-(1-28) is the smallest reported fragment to fully activate cAMP. Neugebauer et al. (1995), [0013] Biochem. 34: 8835-42. However, hPTH-(1-28) was initially reported to have no osteogenic effects in OVX rats. Miller et al. (1997), J. Bone Min. Res. 12: S320 (Abstract). Recently, a very high dose of PTH-(1-28) (1,000 μg/kg/day) was shown to be anabolic in OVX rats, whereas 200 μg/kg/day was ineffective. Whitfield et al. (2000), J. Bone Min. Res. 15: 964-70. The diminished or absent anabolic effects of some truncated PTH fragments has been attributed to rapid clearance in vivo. Rixon et al. (1994).
  • Recombinant and modified proteins are an emerging class of therapeutic agents. Useful modifications of protein therapeutic agents include combination with the “Fc” domain of an antibody and linkage to polymers such as polyethylene glycol (PEG) and dextran. Such modifications are discussed in detail in a patent application entitled, “Modified Peptides as Therapeutic Agents,” U.S. Ser. No. 09/428,082, PCT appl. no. WO 99/25044, which is hereby incorporated by reference in its entirety. [0014]
  • A much different approach to development of therapeutic agents is peptide library screening. The interaction of a protein ligand with its receptor often takes place at a relatively large interface. However, as demonstrated for human growth hormone and its receptor, only a few key residues at the interface contribute to most of the binding energy. Clackson et al. (1995), [0015] Science 267: 383-6. The bulk of the protein ligand merely displays the binding epitopes in the right topology or serves functions unrelated to binding. Thus, molecules of only “peptide” length (2 to 40 amino acids) can bind to the receptor protein of a given large protein ligand. Such peptides may mimic the bioactivity of the large protein ligand (“peptide agonists”) or, through competitive binding, inhibit the bioactivity of the large protein ligand (“peptide antagonists”).
  • Phage display peptide libraries have emerged as a powerful method in identifying such peptide agonists and antagonists. See, for example, Scott et al. (1990), [0016] Science 249: 386; Devlin et al. (1990), Science 249: 404; U.S. Pat. No. 5,223,409, issued Jun. 29, 1993; U.S. Pat. No. 5,733,731, issued Mar. 31, 1998; U.S. Pat. No. 5,498,530, issued Mar. 12, 1996; U.S. Pat. No. 5,432,018, issued Jul. 11, 1995; U.S. Pat. No. 5,338,665, issued Aug. 16, 1994; U.S. Pat. No. 5,922,545, issued Jul. 13, 1999; WO 96/40987, published Dec. 19, 1996; and WO 98/15833, published Apr. 16, 1998 (each of which is incorporated by reference in its entirety). In such libraries, random peptide sequences are displayed by fusion with coat proteins of filamentous phage. Typically, the displayed peptides are affinity-eluted against an antibody-immobilized extracellular domain of a receptor. The retained phages may be enriched by successive rounds of affinity purification and repropagation. The best binding peptides may be sequenced to identify key residues within one or more structurally related families of peptides. See, e.g., Cwirla et al. (1997), Science 276: 1696-9, in which two distinct families were identified. The peptide sequences may also suggest which residues may be safely replaced by alanine scanning or by mutagenesis at the DNA level. Mutagenesis libraries may be created and screened to further optimize the sequence of the best binders. Lowman (1997), Ann. Rev. Biophys. Biomol. Struct. 26: 401-24.
  • Structural analysis of protein-protein interaction may also be used to suggest peptides that mimic the binding activity of large protein ligands. In such an analysis, the crystal structure may suggest the identity and relative orientation of critical residues of the large protein ligand, from which a peptide may be designed. See, e.g., Takasaki et al. (1997), [0017] Nature Biotech. 15: 1266-70. These analytical methods may also be used to investigate the interaction between a receptor protein and peptides selected by phage display, which may suggest further modification of the peptides to increase binding affinity.
  • Other methods compete with phage display in peptide research. A peptide library can be fused to the carboxyl terminus of the lac repressor and expressed in [0018] E. coli. Another E. coli-based method allows display on the cell's outer membrane by fusion with a peptidoglycan-associated lipoprotein (PAL). Hereinafter, these and related methods are collectively referred to as “E. coli display.” In another method, translation of random RNA is halted prior to ribosome release, resulting in a library of polypeptides with their associated RNA still attached. Hereinafter, this and related methods are collectively referred to as “ribosome display.” Other methods employ peptides linked to RNA; for example, PROfusion technology, Phylos, Inc. See, for example, Roberts & Szostak (1997), Proc. Natl. Acad. Sci. USA, 94: 12297-303. Hereinafter, this and related methods are collectively referred to as “RNA-peptide screening.” Chemically derived peptide libraries have been developed in which peptides are immobilized on stable, non-biological materials, such as polyethylene rods or solvent-permeable resins. Another chemically derived peptide library uses photolithography to scan peptides immobilized on glass slides. Hereinafter, these and related methods are collectively referred to as “chemical-peptide screening.” Chemical-peptide screening may be advantageous in that it allows use of D-amino acids and other unnatural analogues, as well as non-peptide elements. Both biological and chemical methods are reviewed in Wells & Lowman (1992), Curr. Opin. Biotechnol. 3: 355-62. Conceptually, one may discover peptide mimetics of any protein using phage display, RNA-peptide screening, and the other methods mentioned above.
  • SUMMARY OF THE INVENTION
  • The present invention concerns therapeutic agents that modulate the activity of PTH and PTHrP. In accordance with the present invention, modulators of PTH and PTHrP comprise: [0019]
  • a) a PTH/PTHrP modulating domain, preferably the amino acid sequence of PTH/PTHrP modulating domains of PTH and/or PTHrP, or sequences derived therefrom by phage display, RNA-peptide screening, or the other techniques mentioned above; and [0020]
  • b) a vehicle, such as a polymer (e.g., PEG or dextran) or an Fc domain, which is preferred; [0021]
  • wherein the vehicle is covalently attached to the carboxyl terminus of the PTH/PTHrP modulating domain. The preferred vehicle is an Fc domain, and the preferred Fc domain is an IgG Fc domain. Preferred PTH/PTHrP modulating domains comprise the PTH and PTHrP-derived amino acid sequences described hereinafter. Other PTH/PTHrP modulating domains can be generated by phage display, RNA-peptide screening and the other techniques mentioned herein. Such peptides typically will be antagonists of both PTH and PTHrP, although such techniques can be used to generate peptide sequences that serve as selective inhibitors (e.g., inhibitors of PTH but not PTHrP). [0022]
  • Further in accordance with the present invention is a process for making PTH and PTHrP modulators, which comprises: [0023]
  • a) selecting at least one peptide that binds to the PTH-1 or PTH-2 receptor; and [0024]
  • b) covalently linking said peptide to a vehicle. [0025]
  • The preferred vehicle is an Fc domain. Step (a) is preferably carried out by selection from the peptide sequences in Tables 1 and 2 hereinafter or from phage display, RNA-peptide screening, or the other techniques mentioned herein. [0026]
  • The compounds of this invention may be prepared by standard synthetic methods, recombinant DNA techniques, or any other methods of preparing peptides and fusion proteins. Compounds of this invention that encompass non-peptide portions may be synthesized by standard organic chemistry reactions, in addition to standard peptide chemistry reactions when applicable. [0027]
  • The primary use contemplated for the compounds of this invention is as therapeutic or prophylactic agents. The vehicle-linked peptide may have activity comparable to—or even greater than—the natural ligand mimicked by the peptide. [0028]
  • The compounds of this invention may be used for therapeutic or prophylactic purposes by formulating them with appropriate pharmaceutical carrier materials and administering an effective amount to a patient, such as a human (or other mammal) in need thereof. Other related aspects are also included in the instant invention. [0029]
  • Of particular interest in the present invention are molecules comprising PTH/PTHRP modulating domains having a shortened PTH C-terminal sequence, such as PTH-(1-28) or (1-34). The prior art shows no anabolic studies using sustained duration delivery of such C-terminally truncated PTH fragments. Although the art does not suggest it, molecules comprising smaller fragments such as PTH-(1-30)-Fc can be anabolic on their own. Despite their weak agonism towards PLC (see Background of the Invention), hPTH-(1-30) is nearly as effective at cAMP stimulation as is hPTH-(1-34). While not wanting to be constrained by theory, the inventors note that the anabolic properties of PTH fragments may be selectively related to their cAMP activation, rather than PLC activation, so that PTH fragments with reduced receptor affinity will have a favorable anabolic profile. It is possible that continuous exposure to truncated PTH fragments would have a different, and more favorable effect on bone compared to continuous exposure to PTH-(1-34) or PTH-(1-84) that has been demonstrated in humans by Fraher et al. (1999). [0030]
  • Numerous additional aspects and advantages of the present invention will become apparent upon consideration of the figures and detailed description of the invention.[0031]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows exemplary Fc dimers that may be derived from an IgG1 antibody. “Fc” in the figure represents any of the Fc variants within the meaning of “Fc domain” herein. “X[0032] 1” and “X2” represent peptides or linker-peptide combinations as defined hereinafter. The specific dimers are as follows:
  • A: Single disulfide-bonded dimers. IgG1 antibodies typically have two disulfide bonds at the hinge region between the constant and variable domains. The Fc domain in FIG. 1A may be formed by truncation between the two disulfide bond sites or by substitution of a cysteinyl residue with an unreactive residue (e.g., alanyl). In FIG. 1A, the Fc domain is linked at the C-terminus of the peptide. [0033]
  • B: Doubly disulfide-bonded dimers. This Fc domain may be formed by truncation of the parent antibody to retain both cysteinyl residues in the Fc domain chains or by expression from a construct including a sequence encoding such an Fc domain. In FIG. 1B, the Fc domain is linked at the C-terminus of the peptide. [0034]
  • C: Noncovalent dimers. This Fc domain may be formed by elimination of the cysteinyl residues by either truncation or substitution. One may desire to eliminate the cysteinyl residues to avoid impurities formed by reaction of the cysteinyl residue with cysteinyl residues of other proteins present in the host cell. The noncovalent bonding of the Fc domains is sufficient to hold together the dimer. Other dimers may be formed by using Fc domains derived from different types of antibodies (e.g., IgG2, IgM). [0035]
  • FIG. 2 shows the structure of additional compounds of the invention. FIG. 2A shows a single chain molecule and may also represent the DNA construct for the molecule. FIG. 2B shows a dimer in which the linker-peptide portion is present on only one chain of the dimer. FIG. 2C shows a dimer having the peptide portion on both chains. The dimer of FIG. 2C will form spontaneously in certain host cells upon expression of a DNA construct encoding the single chain as shown in FIG. 3. In other host cells, the cells could be placed in conditions favoring formation of dimers or the dimers can be formed in vitro. [0036]
  • FIG. 3 shows exemplary nucleic acid and amino acid sequences (SEQ ID NOS: 1 and 2, respectively) of human IgG1 Fc that may be used in this invention. [0037]
  • FIG. 4 shows the calcemic response of normal mice to PTH-(1-34) and to PTH-(1-34)-Fc. Mice were challenged with vehicle (PBS, —X—), or with PTH-(1-34) (open symbols) or with PTH-(1-34)-Fc (closed symbols). Doses were 156 nmol/kg (circles), 469 nmol/kg (triangles) or 1,560 nmol/kg (squares). Data represent group means, n=6 mice/group. [0038]
  • FIG. 5 shows that [Asn10,Leu11]PTHrP-(7-34)-Fc inhibits the calcemic response of normal mice to PTHrP. Normal male mice were injected SC with vehicle (PBS, circles) or with human PTHrP-(1-34) at 0.5 mg/kg (squares). PTHrP-challenged mice were then immediately injected SC with [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 mg/kg (triangles) or 30 mg/kg (diamonds). Data represent group means, with an n of 6 mice/group. [0039]
  • FIG. 6 shows the effect of [Asn10,Leu11]PTHrP-(7-34)-Fc on chronic hypercalcemia induced by PTH-(1-34)-Fc. Normal male mice were challenged once by SC injection with PTH-(1-34)-Fc (30 mg/kg) (open circles), or with vehicle (PBS, open squares). Some PTH-(1-34)-Fc-challenged mice were treated once, at the time of challenge, with [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 (closed triangle), 30 (closed circle), or 100 mg/kg (closed square). All doses of [Asn10,Leu11]PTHrP-(7-34)-Fc caused a significant suppression of PTH-(1-34)-Fc-mediated hypercalcemia. Data represent means±SEM, n=5 mice/group. [0040]
  • FIG. 7 shows cAMP accumulation in ROS 17/2.8 rat osteoblast-like cells. Cultures were treated with the phosphodiesterase inhibitor IBMX and then challenged for 15 minutes with various PTH fragments. cAMP was measured by ELISA. [0041]
  • FIG. 8 shows the effects of single treatments on clinical chemistry. Peripheral blood was obtained daily for 3 days following single subcutaneous injections of the indicated compounds. FIG. 8A shows total serum calcium; FIG. 8B, alkaline phosphatase (AP), a marker of osteoblast activity; FIG. 8C, tartrate-resistant acid phosphatase (TRAP), a marker of osteoclast activity, and FIG. 8D, AP:TRAP ratio, an index of relative osteoblas: osteoclast activity. [0042]
  • FIG. 9 shows the effects of PTH constructs on bone mineral density. Peripheral quantitative computed tomography (pQCT) was performed on the proximal tibial metaphysis of mice on [0043] day 15, after injections of PTH constructs on day 0, 5 and 10.
  • FIG. 10 shows the effect of twice-weekly PTH-(1-34)-Fc versus daily PTH-(1-34) on tibial, trabecular, and cortical bone mineral density (BMD). Daily PTH [PTH-(1-34)] was given at 80 μg/kg/day (20 nmol/kg/day). [0044]
  • FIG. 11 shows the effects of twice-weekly treatment on BMD and serum calcium in aged ovariectomized (OVX) rats. Eleven months after OVX, rats were treated twice per week with phosphate-buffered saline (PBS, vehicle) or with APD (0.5 mg/kg) or with PTH-(1-34)-Fc (50 nmol/kg). DEXA was performed weekly. Blood was drawn 24 hours after the second weekly injection, when the calcemic effects of PTH-Fc are typically maximal. [0045]
  • FIG. 12 shows the effect of a single subcutaneous injection of PTH-(1-34)-Fc into OVX cynomologus monkeys. Monkeys were injected with PTH-(1-34)-Fc at doses of 1-30 μg/kg (n=1/group) or 100-1000 μg/kg (n=2/group). Serum was analyzed for total calcium. The dotted line indicates the threshold for hypercalcemia, based on an elevation of calcium greater than three standard deviations above the normal mean, on two or more consecutive timepoints.[0046]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Definition of Terms [0047]
  • The terms used throughout this specification are defined as follows, unless otherwise limited in specific instances. [0048]
  • The term “comprising” means that a compound may include additional amino acids on either or both of the N— or C-termini of the given sequence. Of course, these additional amino acids should not significantly interfere with the activity of the compound. [0049]
  • The term “acidic residue” refers to amino acid residues in D- or L-form having sidechains comprising acidic groups. Exemplary acidic residues include D and E. [0050]
  • The term “aromatic residue” refers to amino acid residues in D- or L-form having sidechains comprising aromatic groups. Exemplary aromatic residues include F, Y, and W. [0051]
  • The term “basic residue” refers to amino acid residues in D- or L-form having sidechains comprising basic groups. Exemplary basic residues include H, K, and R. [0052]
  • The terms “hydrophilic residue” and “Haa” refer to amino acid residues in D- or L-form having sidechains comprising at least one hydrophilic functional group or polar group. Exemplary hydrophilic residues include C, D, E, H, K, N, Q, R, S, and T. [0053]
  • The terms “lipophilic residue” and “Laa” refer to amino acid residues in D- or L-form having sidechains comprising uncharged, aliphatic or aromatic groups. Exemplary lipophilic sidechains include F, I, L, M, V, W, and Y. Alanine (A) is amphiphilic—it is capable of acting as a hydrophilic or lipophilic residue. Alanine, therefore, is included within the definition of both “lipophilic residue” and “hydrophilic residue.”[0054]
  • The term “nonfunctional residue” refers to amino acid residues in D- or L-form having sidechains that lack acidic, basic, or aromatic groups. Exemplary nonfunctional amino acid residues include M, G, A, V, I, L and norleucine (Nle). [0055]
  • The term “vehicle” refers to a molecule that prevents degradation and/or increases half-life, reduces toxicity, reduces immunogenicity, or increases biological activity of a therapeutic protein. Exemplary vehicles include an Fc domain (which is preferred) as well as a linear polymer (e.g., polyethylene glycol (PEG), polylysine, dextran, etc.); a branched-chain polymer (see, for example, U.S. Pat. No. 4,289,872 to Denkenwalter et al., issued Sep. 15, 1981; U.S. Pat. No. 5,229,490 to Tam, issued Jul. 20, 1993; WO 93/21259 by Frechet et al., published 28 Oct. 1993); a lipid; a cholesterol group (such as a steroid); a carbohydrate or oligosaccharide (e.g., dextran); human serum albumin (HSA) and related molecules; transtheratin (TTR) and related molecules; or any natural or synthetic protein, polypeptide or peptide that binds to a salvage receptor. Vehicles are further described hereinafter. [0056]
  • The term “native Fc” refers to molecule or sequence comprising the sequence of a non-antigen-binding fragment resulting from digestion of whole antibody, whether in monomeric or multimeric form. The original immunoglobulin source of the native Fc is preferably of human origin and may be any of the immunoglobulins, although IgG1 and IgG2 are preferred. Native Fc's are made up of monomeric polypeptides that may be linked into dimeric or multimeric forms by covalent (i.e., disulfide bonds) and non-covalent association. The number of intermolecular disulfide bonds between monomeric subunits of native Fc molecules ranges from 1 to 4 depending on class (e.g., IgG, IgA, IgE) or subclass (e.g., IgG1, IgG2, IgG3, IgA1, IgGA2). One example of a native Fc is a disulfide-bonded dimer resulting from papain digestion of an IgG (see Ellison et al. (1982), [0057] Nucleic Acids Res. 10: 4071-9). The term “native Fc” as used herein is generic to the monomeric, dimeric, and multimeric forms.
  • The term “Fc variant” refers to a molecule or sequence that is modified from a native Fc but still comprises a binding site for the salvage receptor, FcRn. International applications WO 97/34631 (published 25 Sep. 1997) and WO 96/32478 describe exemplary Fc variants, as well as interaction with the salvage receptor, and are hereby incorporated by reference in their entirety. Thus, the term “Fc variant” comprises a molecule or sequence that is humanized from a non-human native Fc. Furthermore, a native Fc comprises sites that may be removed because they provide structural features or biological activity that are not required for the fusion molecules of the present invention. Thus, the term “Fc variant” comprises a molecule or sequence that lacks one or more native Fc sites or residues that affect or are involved in (1) disulfide bond formation, (2) incompatibility with a selected host cell (3) N-terminal heterogeneity upon expression in a selected host cell, (4) glycosylation, (5) interaction with complement, (6) binding to an Fc receptor other than a salvage receptor, or (7) antibody-dependent cellular cytotoxicity (ADCC). Fc variants are described in further detail hereinafter. [0058]
  • The term “Fc domain” encompasses native Fc and Fc variant molecules and sequences as defined above. As with Fc variants and native Fc's, the term “Fc domain” includes molecules in monomeric or multimeric form, whether digested from whole antibody or produced by other means. [0059]
  • The term “multimer” as applied to Fc domains or molecules comprising Fc domains refers to molecules having two or more polypeptide chains associated covalently, noncovalently, or by both covalent and non-covalent interactions. IgG molecules typically form dimers; IgM, pentamers; IgD, dimers; and IgA, monomers, dimers, trimers, or tetramers. Multimers may be formed by exploiting the sequence and resulting activity of the native Ig source of the Fc or by derivatizing (as defined below) such a native Fc. [0060]
  • The term “dimer” as applied to Fc domains or molecules comprising Fc domains refers to molecules having two polypeptide chains associated covalently or non-covalently. Thus, exemplary dimers within the scope of this invention are as shown in FIGS. 1 and 2. [0061]
  • The terms “derivatizing” and “derivative” or “derivatized” comprise processes and resulting compounds respectively in which (1) the compound has a cyclic portion; for example, cross-linking between cysteinyl residues within the compound; (2) the compound is cross-linked or has a cross-linking site; for example, the compound has a cysteinyl residue and thus forms cross-linked dimers in culture or in vivo; (3) one or more peptidyl linkage is replaced by a non-peptidyl linkage; (4) the N-terminus is replaced by —NRR[0062] 1, NRC(O)R1, —NRC(O)OR1, —NRS(O)2R1, —NHC(O)NHR, a succinimide group, or substituted or unsubstituted benzyloxycarbonyl-NH—, wherein R and R1 and the ring substituents are as defined hereinafter; (5) the C-terminus is replaced by —C(O)R2 or —NR3R4 wherein R2, R3 and R4 are as defined hereinafter; and (6) compounds in which individual amino acid moieties are modified through treatment with agents capable of reacting with selected side chains or terminal residues. Derivatives are further described hereinafter.
  • The term “peptide” refers to molecules of 1 to 85 amino acids, with molecules of 5 to 34 amino acids preferred. Exemplary peptides may comprise the PTH/PTHrP modulating domain of a naturally occurring molecule or comprise randomized sequences. [0063]
  • The term “randomized” as used to refer to peptide sequences refers to fully random sequences (e.g., selected by phage display methods or RNA-peptide screening) and sequences in which one or more residues of a naturally occurring molecule is replaced by an amino acid residue not appearing in that position in the naturally occurring molecule. Exemplary methods for identifying peptide sequences include phage display, [0064] E. coli display, ribosome display, RNA-peptide screening, chemical screening, and the like.
  • The term “PTH/PTHrP modulating domain” refers to any amino acid sequence that binds to the PTH-1 receptor and/or the PTH-2 receptor and comprises naturally occurring sequences or randomized sequences. Exemplary PTH/PTHrP modulating domains can be identified or derived as described in the references listed for Tables 1 and 2, which are hereby incorporated by reference in their entirety. [0065]
  • The term “PTH agonist” refers to a molecule that binds to PTH-1 or PTH-2 receptor and increases or decreases one or more PTH activity assay parameters as does full-length native human parathyroid hormone. An exemplary PTH activity assay is disclosed in Example 1. [0066]
  • The term “PTH antagonist” refers to a molecule that binds to PTH-1 or PTH-2 receptor and blocks or prevents the normal effect on those parameters by full length native human parathyroid hormone. An exemplary PTH activity assay is disclosed in Example 2. [0067]
  • The term “bone resorption inhibitor” refers to such molecules as determined by the assays of Examples 4 and 11 of WO 97/23614:, which is hereby incorporated by reference in its entirety. Exemplary bone resorption inhibitors include OPG and OPG-L antibody, which are described in WO 97/23614 and WO98/46751, respectively, which are hereby incorporated by reference in their entirety. [0068]
  • Additionally, physiologically acceptable salts of the compounds of this invention are also encompassed herein. By “physiologically acceptable salts” is meant any salts that are known or later discovered to be pharmaceutically acceptable. Some specific examples are: acetate; trifluoroacetate; hydrohalides, such as hydrochloride and hydrobromide; sulfate; citrate; tartrate; glycolate; and oxalate. [0069]
  • Structure of Compounds [0070]
  • In General. PTH and PTHrP receptor binding amino acid sequences are described in Tables 1 and 2. Other information on PTH and PTHrP is found in Mannstadt et al. (1999), [0071] Am. J. Physiol. 277. 5Pt 2: F665-75; and Gardella (1996), J. Biol. Chem. 271 (33): 19888-93. Each of these references is hereby incorporated by reference in its entirety.
  • From the foregoing sequences, the present inventors identified in particular preferred sequences derived from PTH and PTHrP. These sequences can be randomized through the techniques mentioned above by which one or more amino acids may be changed while maintaining or even improving the binding affinity of the peptide. [0072]
  • In the compositions of matter prepared in accordance with this invention, the peptide may be attached to the vehicle through the peptide's C-terminus. Thus, the vehicle-peptide molecules of this invention may be described by the following formula I: [0073]
  • P1-(L1)a-F1
  • and multimers thereof, wherein: [0074]
  • F[0075] 1 is a vehicle (preferably an Fc domain) and is attached at the C-terminus of P1-(L1)a;
  • P[0076] 1 is a sequence of a PTH/PTHrP modulating domain;
  • L[0077] 1 is a linker; and
  • a is 0 or 1. [0078]
  • Peptides. Any number of peptides may be used in conjunction with the present invention. Peptides may comprise part of the sequence of naturally occurring proteins, may be randomized sequences derived from the sequence of the naturally occurring proteins, or may be wholly randomized sequences. Phage display and RNA-peptide screening, in particular, are useful in generating peptides for use in the present invention. [0079]
  • A PTH/PTHrP modulating domain sequence particularly of interest is of the formula II [0080]
    XNX8HX10X11X12KX14X15X16X17X18X19 (SEQ ID NO: 3)
    RX21X22X23X24X25X26X27X28XC
  • wherein: [0081]
  • X[0082] N is absent or is X3X4X5X6X7, X2X3X4X5X6X7, X1X2X3X4X5X6X7, or YX1X2X3X4X5X6X7;
  • X[0083] 1 is an amino acid residue (nonfunctional, hydrophilic or aromatic residue preferred; A, S or Y preferred);
  • X[0084] 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • X[0085] 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • X[0086] 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • X[0087] 5 is an amino acid residue (nonfunctional or basic residue preferred, H or I most preferred);
  • X[0088] 6 is an amino acid residue (acidic or hydrophilic residue preferred, Q or E most preferred);
  • X[0089] 7 is an amino acid residue (nonfunctional or aromatic residue preferred, L or F most preferred);
  • X[0090] 8 is an amino acid residue (nonfunctional residue preferred, M or Nle most preferred);
  • X[0091] 10 is an amino acid residue (an acidic or hydrophilic residue preferred, N or D most preferred);
  • X[0092] 11 is an amino acid residue (nonfunctional or basic residue preferred, L, R, or K most preferred);
  • X[0093] 12 is an amino acid residue (nonfunctional or aromatic residue preferred, G, F, or W most preferred);
  • X[0094] 14 is an amino acid residue (basic or hydrophilic residue preferred, H or S most preferred);
  • X[0095] 15 is an amino acid residue (nonfunctional residue preferred, with L or I most preferred);
  • X[0096] 16 is an amino acid residue (nonfunctional or hydrophilic residue preferred, Q, N, S, or A most preferred);
  • X[0097] 17 is an amino acid residue (acidic, hydrophilic, or nonfunctional residue preferred; S, D, or L most preferred);
  • X[0098] 18 is an amino acid residue (nonfunctional residue preferred, M, L, V or Nle most preferred);
  • X[0099] 19 is an amino acid residue (acidic or basic residue preferred, E or R most preferred);
  • X[0100] 21 is an amino acid residue (nonfunctional residue or basic residue preferred; V, M, R, or Nle most preferred);
  • X[0101] 22 is an amino acid residue (hydrophilic, acidic, or aromatic residue preferred, E or F most preferred);
  • X[0102] 23 is an aromatic or lipophilic residue (W or F preferred);
  • X[0103] 24 is a lipophilic residue (L preferred);
  • X[0104] 25 is an amino acid residue (hydrophilic or basic residue preferred, R or H most preferred);
  • X[0105] 26 is an amino acid residue (hydrophilic or basic residue preferred, K or H most preferred);
  • X[0106] 27 is an amino acid residue (lipophilic, basic, or nonfunctional residue preferred, K or L most preferred);
  • X[0107] 28 is an amino acid residue (lipophilic or nonfunctional residue preferred, L or I most preferred);
  • X[0108] C is absent or is X29, X29X30, X29X30X31, X29X30X31X32, X29X30X31X32X33, X29X30X31X32X33X34, X29X30X31X32X33X34X35, or X29X30X31X32X33X34X35X36;
  • X[0109] 29 is an amino acid residue (hydrophilic or nonfunctional residue preferred, Q or A most preferred);
  • X[0110] 30 is an amino acid residue (hydrophilic or acidic residue preferred, D or E most preferred);
  • X[0111] 31 is an amino acid residue (lipophilic or nonfunctional residue preferred, V or I most preferred);
  • X[0112] 32 is an amino acid residue (basic residue preferred, H most preferred);
  • X[0113] 33 is an amino acid residue (hydrophilic residue preferred, N or T most preferred);
  • X[0114] 34 is an amino acid residue (nonfunctional or aromatic residue preferred, A, F or Y most preferred);
  • X[0115] 35 is an amino acid residue (acidic residue preferred, E most preferred); and
  • X[0116] 36 is an amino acid residue (aromatic residue preferred, Y most preferred).
  • A preferred PTH/PTHrP modulating domain sequence formula is III [0117]
    JNJ7J8HNLJ12KHLJ16SJ18 (SEQ ID NO: 4)
    J19RJ21EWLRKKLJC
  • wherein: [0118]
  • J[0119] N is absent or is selected from J1J2J3J4J5J6, J2J3J4J5J6, J3J4J5J6;
  • J[0120] 1 is an amino acid residue (nonfunctional, hydrophilic, or aromatic residue preferred; A, S or Y most preferred);
  • J[0121] 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • J[0122] 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • J[0123] 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • J[0124] 5 is an amino acid residue (nonfunctional residue preferred, I most preferred);
  • J[0125] 6 is an amino acid residue (basic residue preferred, Q preferred);
  • J[0126] 7 is an amino acid residue (nonfunctional or aromatic residue preferred, L or F most preferred);
  • J[0127] 8 is an amino acid residue (nonfunctional residue preferred, M or Nle most preferred);
  • J[0128] 12 is an amino acid residue (nonfunctional or aromatic residue preferred, G or W most preferred);
  • J[0129] 16 is an amino acid residue (nonfunctional or hydrophilic residue preferred, N, S, or A most preferred);
  • J[0130] 18 is an amino acid residue (nonfunctional residue preferred, M, Nle, L, or V most preferred);
  • J[0131] 19 is an acidic or basic residue (E or R preferred);
  • J[0132] 21 is an amino acid residue (nonfunctional residue preferred, V, M, or Nle most preferred);
  • J[0133] C is absent or is J29, J29J30, J29J30J31, J29J30J31J32, J29J30J31J32J33, or J29J30J31J32J33J34;
  • J[0134] 29 is an amino acid residue (hydrophilic or nonfunctional residue preferred, Q or A most preferred);
  • J[0135] 30 is an amino acid residue (hydrophilic or acidic residue preferred, D or E most preferred);
  • J[0136] 31 is an amino acid residue (lipophilic or nonfunctional residue preferred, V or I most preferred);
  • J[0137] 32 is an amino acid residue (basic residue preferred, H most preferred);
  • J[0138] 33 is an amino acid residue (acidic residue preferred, N most preferred);
  • J[0139] 34 is an amino acid residue (aromatic residue preferred, F or Y most preferred).
  • From the formula of SEQ ID NO: 4, peptides appearing in Table 1 below are most preferred. [0140]
  • Another preferred PTH/PTHrP modulating domain sequence is IV [0141]
    ONLHO10O11O12KSIO15 (SEQ ID NO: 5)
    O16LRRRFO23LHHLIOC
  • wherein: [0142]
  • O[0143] N is absent or is YO1O2O3O4O5O6O7, O1O2O3O4O5O6O7, O2O3O4O5O6O7, O3O4O5O6O7, O4O5O6O7, O5 6O7, O6O7, or O7;
  • O[0144] 1 is an amino acid residue (nonfunctional residue preferred, A most preferred);
  • O[0145] 2 is an amino acid residue (nonfunctional residue preferred, V most preferred);
  • O[0146] 3 is an amino acid residue (hydrophilic residue preferred, S most preferred);
  • O[0147] 4 is an amino acid residue (acidic residue preferred, E most preferred);
  • O[0148] 5 is an amino acid residue (basic or nonfunctional residue preferred, H or I preferred);
  • O[0149] 6 is an amino acid residue (hydrophilic residue preferred, Q most preferred);
  • O[0150] 7 is an amino acid residue (nonfunctional residue preferred, L most preferred);
  • O[0151] 10 is an amino acid residue (acidic or hydrophilic residue preferred, N or D most preferred);
  • O[0152] 11 is an amino acid residue (basic or nonfunctional residue preferred, K or L most preferred);
  • O[0153] 12 is an amino acid residue (aromatic or nonfunctional residue preferred, G, F, or W most preferred);
  • O[0154] 15 is an amino acid residue (hydrophilic or nonfunctional residue preferred, I or S most preferred);
  • O[0155] 16 is an amino acid residue (hydrophilic residue preferred, Q or N most preferred);
  • O[0156] 17 is an amino acid residue (acidic or nonfunctional residue preferred, D or L most preferred);
  • O[0157] 23 is an amino acid residue (aromatic residue preferred, with F or W most preferred);
  • O[0158] C is absent or is O29, O29O30, O29O30O31, O29O30O31O32, O29O30O31O32O33, O29O30O31O32O33O34, O29O30O31O32O33O34O35, or O29O30O31O32O33O34O35O36; and
  • O[0159] 29 through O36 are each independently amino acid residues.
  • From the formula of SEQ ID NO: 5, peptides appearing in Table 2 below are most preferred. [0160]
  • Exemplary peptide sequences for this invention appear in Tables 1 and 2 below. These peptides may be prepared as described in the cited references or in U.S. Pat. Nos. 4,423,037, 4,968,669, 5,001,22, and 6,051,686, each of which is hereby incorporated by reference in its entirety. Molecules of this invention incorporating these peptide sequences may be prepared by methods known in the art. Single letter amino acid abbreviations are used. Any of these peptides may be linked in tandem (i.e., sequentially), with or without linkers. Any peptide containing a cysteinyl residue may be cross-linked with another Cys-containing peptide, either or both of which may be linked to a vehicle. Any peptide having more than one Cys residue may form an intrapeptide disulfide bond, as well. Any of these peptides may be derivatized as described hereinafter. [0161]
    TABLE 1
    PTH/PTHrP modulating domains based on PTH
    SEQ
    ID
    Description Sequence NO:
    human PTH(1-84)1 SVSEIQLMHNLGKHLNSMERVE 10
    WLRKKLQDVHNFVALGAPLAPR
    DAGSQRPRKKEDNVLVESHEKS
    LGEADKADVNVLTKAKSQ
    rat PTH(1-84)2 AVSEIQLMHNLGKHLASVERMQ 11
    WLRKKLQDVHNFVSLGVQMAAR
    EGSYQRPTKKEDNVLVDGNSKS
    LGEGDKADVDVLVKAKSQ
    human PTH3(7-84) LMHNLGKHLNSMERVEWLRKKL 12
    QDVHNFVALGAPLAPRDAGSQR
    PRKKEDNVLVESHEKSLGEADK
    ADVNVLTKAKSQ
    human PTH(1-44)3 SVSEIQLMHNLGKHLNSMERVE 13
    WLRKKLQDVHNFVALGAPLAPR
    human PTH(1-38)3 SVSEIQLMHNLGKHLNSMERVE 14
    WLRKKLQDVHNFVALG
    human PTH(2-38)3 VSEIQLMHNLGKHLNSMERVEW 15
    LRKKLQDVHNFVALG
    human PTH(1-34)4 SVSEIQLMHNLGKHLNSMERVE 16
    WLRKKLQDVHNF
    [Arg11] human PTH(1-34) SVSEIQLMHNRGKHLNSMERVE 17
    WLRKKLQDVHNF
    [Lys11] human PTH(1-34) SVSEIQLMHNKGKHLNSMERVE 18
    WLRKKLQDVHNF
    [Arg19] human PTH(1-34) SVSEIQLMHNLGKHLNSMRRVE 19
    WLRKKLQDVHNF
    [Tyr1] human PTH(1-34)3 YVSEIQLMHNLGKHLNSMERVE 20
    WLRKKLQDVHNF
    [Leu(8, 18), Tyr34] SVSEIQLLHNLGKHLNSLERVE 21
    human PTH(1-34)3 WLRKKLQDVHNY
    bovine PTH(1-34)5 AVSEIQFMHNLGKHLSSMERVE 22
    WLRKKLQDVHNF
    [Leu(8, 18), Tyr34] AVSEIQFLHNLGKHLSSLERVE 23
    bovine (1-34)6 WLRKKLQDVHNY
    porcine PTH(1-34)3 SVSEIQLMHNLGKHLSSLERVE 24
    WLRKKLQDVHNF
    rat PTH(1-34)3 AVSEIQLMHNLGKHLASVERMQ 25
    WLRKKLQDVHNF
    [Leu(8, 21), Tyr34] rat AVSEIQLLHNLGKHLASVERLQ 26
    PTH(1-34)3 WLRKKLQDVHNY
    human PTH(1-31)7 SVSEIQLMHNLGKHLNSMERVE 27
    WLRKKLQDV
    [Leu27] human PTH(1-31)8 SVSEIQLMHNLGKHLNSMERVE 28
    WLRKLLQDV
    [Leu(8, 18) Tyr34] SEIQLLHNLGKHLNSLERVEWL 29
    PTH(3-34)9 RKKLQDVHNY
    bovine PTH(3-34)10 SEIQFMHNLGKHLSSMERVEWL 30
    RKKLQDVHNF
    [Leu(8, 18), Tyr34] SEIQFLHNLGKHLSSLERVEWL 31
    bovine PTH(3-34)11 RKKLQDVHNY
    human PTH(7-34)12 LMHNLGKHLNSMERVEWLRKKL 32
    QDVHNF
    [Leu(8, 18) Tyr34] LLHNLGKHLNSLERVEWLRKKL 33
    human PTH(7-34)9 QDVHNY
    bovine PTH(7-34)13 FMHNLGKHLSSMERVEWLRKKL 34
    QDVHNF
    [Tyr34] bovine FMHNLGKHLSSMERVEWLRKKL 35
    PTH(7-34)14 QDVHNY
    [Leu(8, 18), Tyr34] FLHNLGKHLSSLERVEWLRKKL 36
    bovine PTH(7-34)15 QDVHNY
    [Leu(8, 18), Trp12, FLHNLWKHLSSLERVEWLRKKL 37
    Tyr34] bovine QDVHNY
    PTH(7-34)16
    [D-Trp12, Tyr34] FMHNL-D-Trp-KHLSSMERVE 38
    bovine PTH(7-34)17 WLRKKLQDVHNY
    human PTH(1-30) SVSEIQLMHNLGKHLNSMERVE 39
    WLRKKLQD
    [Arg11] human PTH(1-30) SVSEIQLMHNRGKHLNSMERVE 40
    WLRKKLQD
    [Lys11] human PTH(1-30) SVSEIQLMHNKGKHLNSMERVE 41
    WLRKKLQD
    [Arg19] human PTH(1-30) SVSEIQLMHNLGKHLNSMRRVE 42
    WLRKKLQD
    [Tyr1] human PTH(1-30) YVSEIQLMHNLGKHLNSMERVE 43
    WLRKKLQD
    [Leu(8, 18)] human SVSEIQLLHNLGKHLNSLERVE 44
    PTH(1-30) WLRKKLQD
    bovine PTH(1-30) AVSEIQFMHNLGKHLSSMERVE 45
    WLRKKLQD
    [Leu(8, 18)] bovine AVSEIQFLHNLGKHLSSLERVE 46
    PTH(1-30) WLRKKLQD
    porcine PTH(1-30) SVSEIQLMHNLGKHLSSLERVE 47
    WLRKKLQD
    rat PTH(1-30) AVSEIQLMHNLGKHLASVERMQ 48
    WLRKKLQD
    [Leu(8, 21), Tyr34] rat AVSEIQLLHNLGKHLASVERLQ 49
    PTH(1-30) WLRKKLQD
    [Leu27] human PTH(1-30) SVSEIQLMHNLGKHLNSMERVE 50
    WLRKLLQD
    human PTH(1-29) SVSEIQLMHNLGKHLNSMERVE 51
    WLRKKLQ
    human PTH(1-28) SVSEIQLMHNLGKHLNSMERVE 52
    WLRKKL
    [Leu(8, 18)] PTH(3-30) SEIQLLHNLGKHLNSLERVEWL 53
    RKKLQD
    bovine PTH(3-30) SEIQFMHNLGKHLSSMERVEWL 54
    RKKLQD
    [Leu(8, 18)] bovine SEIQFLHNLGKHLSSLERVEWL 55
    PTH(3-30) RKKLQD
    human PTH(7-30) LMHNLGKHLNSMERVEWLRKKL 56
    QD
    [Leu(8, 18)] human LLHNLGKHLNSLERVEWLRKKL 57
    PTH(7-30) QD
    bovine PTH(7-30) FMHNLGKHLSSMERVEWLRKKL 58
    QD
    [Leu(8, 18)] bovine FLHNLGKHLSSLERVEWLRKKL 59
    PTH(7-30) QD
    [Leu(8, 18), Trp12] FLHNLWKHLSSLERVEWLRKKL 60
    bovine PTH(7-30) QD
    [D-Trp12] bovine FMHNL-D-Trp-KHLSSMERVE 61
    PTH(7-30) WLRKKLQD
  • [0162]
    TABLE 2
    PTH/PTHrP modulating domains based on PTHrP
    SEQ
    ID
    Description Sequence NO:
    human PTHrP(1-86)3 AVSEHQLLHDKGKSIQDL  62
    RRRFFLHHLIAEIHTAEI
    RATSEVSPNSKPSPNTKN
    HPVRFGSDDEGRYLTQET
    NKVETYKEQPLKTP
    human PTHrP(1-34)18 AVSEHQLLHDKGKSIQDL  63
    RRRFFLHHLIAEIHTA
    [Tyr36] human PTHrP(1-36)3 AVSEHQLLHDKGKSIQDL  64
    RRRFFLHHLIAEIHTAEY
    [Ile5, Trp23, Tyr36] human AVSEIQLLHDKGKSIQDL  65
    PTHrP(1-36)3 RRRFWLHHLIAEIHTAEY
    Tyr-human PTHrP(1-34)3 YAVSEHQLLHDKGKSIQD  66
    LRRRFFLHHLIAEIHTA
    [Asn10, Leu11, D-Phe12] AVSEHQLLHNL-D-Phe-  67
    human PTHrP(1-34)19 KSIQDLRRRFFLHHLIAE
    IHTA
    PTHrP(7-34)20 LLHDKGKSIQDLRRRFFL  68
    HHLIAEIHTA
    [Asn10, Leu11] human LLHNLGKSIQDLRRRFFL  69
    PTHrP(7-34) HHLIAEIHTA
    [Asn16, Leu17] LLHDKGKSINLLRRRFFL  70
    PTHrP(7-34)21 HHLIAEIHTA
    [Leu11, D-Trp12] human LLHDL-D-Trp-KSIQDL  71
    PTHrP(7-34)22 RRRFFLHHLIAEIHTA
    [Asn10, Leu11, D-Trp12] LLHNL-D-Trp-KSIQDL  72
    PTHrP(7-34)23 RRRFFLHHLIAEIHTA
    [D-Trp12] PTHrP(8-34) LHNL-D-Trp-KSIQDLR  73
    RRFFLHHLIAEIHTA
    [D-Phe12] PTHrP(8-34) LHNL-D-Phe-KSIQDLR  74
    RRFFLHHLIAEIHTA
    [Asn10, Leu11, D-Trp12] LLHNL-D-Trp-KSIQDL  75
    human PTHrP(7-34)20 RRRFFLHHLIAEIHTA
    human PTHrP(1-30) AVSEHQLLHDKGKSIQDL  76
    RRRFFLHHLIAE
    [Ile5, Trp23] human AVSEIQLLHDKGKSIQDL  77
    PTHrP(1-30) RRRFWLHHLIAE
    Tyr-human PTHrP(1-30) YAVSEHQLLHDKGKSIQD  78
    LRRRFFLHHLIAE
    [Asn10, Leu11, D-Phe12] AVSEHQLLHNL-D-Phe-  79
    human PTHrP(1-30) KSIQDLRRRFFLHHLIAE
    PTHrP(7-30) LLHDKGKSIQDLRRRFFL  80
    HHLIAE
    [Asn10, Leu11] human LLHNLGKSIQDLRRRFFL  81
    PTHrP(7-30) HHLIAE
    [Asn16, Leu17] PTHrP(7-30) LLHDKGKSINLLRRRFFL  82
    HHLIAE
    [Leu11, D-Trp12] human LLHDL-D-Trp-KSIQDL  83
    PTHrP(7-30) RRRFFLHHLIAE
    [Asn10, Leu11, D-Trp12] LLHNL-D-Trp-KSIQDL  84
    PTHrP(7-30) RRRFFLHHLIAE
    [D-Trp12] PTHrP(8-30) LHNL-D-Trp-KSIQDLR  85
    RRFFLHHLIAE
    [D-Phe12] PTHrP(8-30) LHNL-D-Phe-KSIQDLR  86
    RRFFLHHLIAE
    [Asn10, Leu11, D-Trp12] LLHNL-D-Trp-KSIQDL  87
    human PTHrP(7-30) RRRFFLHHLIAE
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM  88
    22-31] human PTH(1-34)24 ERVELLEKLLEKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM  89
    22-31] human PTH(1-34)24 ERVELLEKLLKKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM  90
    22-31] human PTH(1-34)25 ERVALAEALAEALHNF
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM  91
    22-31] human PTH(1-34)26 ERVSLLSSLLSSLHNF
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM  92
    22-31] human PTH(1-34)27 ERVAFYDKVAEKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVELL  93
    22-31] human PTH(7-34)24 EKLLEKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVELL  94
    22-31] human PTH(7-34)24 EKLLKKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVALA  95
    22-31] human PTH(7-34)25 EALAEALHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVSLL  96
    22-31] human PTH(7-34)26 SSLLSSLHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVAFY  97
    22-31] human PTH(7-34)27 DKVAEKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL  98
    22-31] human PTHrP(1-34)24 RRRELLEKLLEKLHTA
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL  99
    22-31] human PTHrP(1-34)24 RRRELLEKLLKKLHTA
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 100
    22-31] human PTHrP(1-34)25 RRRALAEALAEALHTA
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 101
    22-31] human PTHrP(1-34)26 RRRSLLSSLLSSLHTA
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 102
    22-31[ human PTHrP(1-34)27 RRRAFYDKVAEKLHTA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRELL 103
    22-31] human PTHrP(7-34)28 EKLLEKLHTA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRELL 104
    22-31] human PTHrP(7-34)24 EKLLKKLHTA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRALA 105
    22-31] human PTHrP(7-34)25 EALAEALHTA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRSLL 106
    22-31] human PTHrP(7-34)26 SSLLSSLHTA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRAFY 107
    22-31] human PTHrP(7-34)27 DKVAEKLHTA
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 108
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLRKLHTA
    22-31] human PTHrP(1-34)29
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 109
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEKLHTS
    22—31] human PTHrP(1-34)30
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 110
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEKLHTAGR
    22-31] human PTHrP(1-34)31 R
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 111
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEKLKEL
    22-31] human PTHrP(1-34)32
    [Lys11, Lys13, Ala19, Arg21, AVSEHQLLHDKGKSIQDL 112
    Haa(Laa Laa Haa Haa)2 Laa ARRELLEKLLEKLHTA
    22-31] human PTHrP(1-34)33
    [Lys11, Lys13, Arg19, Ala21, AVSEHQLLHDKGKSIQDL 113
    Haa(Laa Laa Haa Haa)2 Laa RRAELLEKLLEKLHTA
    22-31] human PTHrP(1-34)34
    [Leu11, Lys13, Arg19, Arg21, AVSEAQLLHDLGKSIQDL 114
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEKLHAL
    22-31] human PTHrP(1-34)35
    [Lys11, Lys13, Arg19, Arg21, AVSEHQLLHDKGKSIQDL 115
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLERLHTA
    22-31] human PTHrP(1-34)36
    [Arg11, Arg13, Arg19, Arg21, AVSEHQLLHDRGRSIQDR 116
    Haa(Laa Laa Haa Haa)2 Laa RRELLERLLERLHTA
    22-31] human PTHrP(1-34)37
    [Arg11, Lys13, Arg19, Arg21, AVSEHQLLHDRGKSIQDL 117
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLKRLHTA
    22-31] human PTHrP(1-34)38
    [Arg11, Arg13, Arg19, Arg21, AVSEHQLLHDRGRSIQDL 118
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLKRLHTA
    22-31] human PTHrP(1-34)39
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 119
    22-31] human PTHrP(1-34)40 RRRALAEALAEALHTA
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 120
    22-31] human PTHrP(1-34)41 RRRSLLSSLLSSLHTA
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 121
    22-31] human PTHrP(1-34)42 RRRAFYDKVAEKLHTA
    Haa(Laa Laa Haa Haa)2 Laa AVSEIQFMHNLGKHLSSM 122
    22-31] human PTHrP(1-34)43 ERVELLEKLLEKLHNY
    Haa(Laa Laa Haa Haa)2 Laa AVSEIQFMHNLGKHLSSM 123
    22-31] human PTHrP(1-34)44 RRRELLEKLLEKLHNY
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM 124
    22-30] human PTH(1-30) ERVELLEKLLEK
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM 125
    22-30] human PTH(1-30) ERVELLEKLLKK
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM 126
    22-30] human PTH(1-30) ERVALAEALAEA
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM 127
    22-30] human PTH(1-30) ERVSLLSSLLSS
    [Haa(Laa Laa Haa Haa)2 Laa SVSEIQLMHNLGKHLNSM 128
    22-30] human PTH(1-34)27 ERVAFYDKVAEKLHNF
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVELL 129
    22-30] human PTH(7-30) EKLLEK
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVELL 130
    22-30] human PTH(7-30) EKLLKK
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVALA 131
    22-30] human PTH(7-30) EALAEA
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVSLL 132
    22-30] human PTH(7-30) SSLLSS
    [Haa(Laa Laa Haa Haa)2 Laa LMHNLGKHLNSMERVAFY 133
    22-30] human PTH(7-30) DKVAEK
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 134
    22-30] human PTHrP(1-30) RRRELLEKLLEK
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 135
    22-30] human PTHrP(1-30) RRRELLEKLLKK
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 136
    22-30] human PTHrP(1-30) RRRALAEALAEA
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 137
    22-30] human PTHrP(1-30) RRRSLLSSLLSS
    [Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 138
    22-30] human PTHrP(1-30) RRRAFYDKVAEK
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRELL 139
    22-30] human PTHrP(7-30) EKLLEK
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRELL 140
    22-30] human PTHrP(7-30) EKLLKK
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRALA 141
    22-30] human PTHrP(7-30) EALAEA
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRRSLL 142
    22-30] human PTHrP(7-30) SSLLSS
    [Haa(Laa Laa Haa Haa)2 Laa LLHDKGKSIQDLRRHAFY 143
    22-30] human PTHrP(7-30) DKVAEK
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 144
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLRK
    22-30] human PTHrP(1-30)
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 145
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEK
    22-30] human PTHrP(1-30)
    ]Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 146
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEKLHT
    22-30] human PTHrP(1-30)
    [Lys11, Lys13; Arg19, Arg21; AVSEHQLLHDKGKSIQDL 147
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEK
    22-30] human PTHrP(1-30)
    [Lys11, Lys13, Ala19, Arg21, AVSEHQLLHDKGKSIQDL 148
    Haa(Laa Laa Haa Haa)2 Laa ARRELLEKLLEK
    22-30] human PTHrP(1-30)
    [Lys11, Lys13, Arg19, Ala21, AVSEHQLLHDKGKSIQDL 149
    Haa(Laa Laa Haa Haa)2 Laa RRAELLEKLLEK
    22-30] human PTHrP(1-30)
    [Leu11, Lys13, Arg19, Arg21, AVSEAQLLHDLGKSIQDL 150
    Haa(Laa Laa Haa Haa)2 Laa RRRELLEKLLEK
    22-30] human PTHrP(1-30)
    [Lys11, Lys13, Arg19, Arg21, AVSEHQLLHDKGKSIQDL 151
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLER
    22-30] human PTHrP(1-30)
    [Arg11, Arg13, Arg19, Arg21, AVSEHQLLHDRGRSIQDR 152
    Haa(Laa Laa Haa Haa)2 Laa RRELLERLLER
    22-30] human PTHrP(1-30)
    [Arg11, Lys13, Arg19, Arg21, AVSEHQLLHDRGKSIQDL 153
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLKR
    22-30] human PTHrP(1-30)
    [Arg11, Arg13, Arg19, Arg21, AVSEHQLLHDRGRSIQDL 154
    Haa(Laa Laa Haa Haa)2 Laa RRRELLERLLKR
    22-30] human PTHrP(1-30)
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 155
    22-30] human PTHrP(1-30) RRREALAEALAEA
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 156
    22-30] human PTHrP(1-30) RRRSLLSSLLSS
    Haa(Laa Laa Haa Haa)2 Laa AVSEHQLLHDKGKSIQDL 157
    22-30] human PTHrP(1-30) RRRAFYDKVAEK
    Haa(Laa Laa Haa Haa)2 Laa AVSEIQFMHNLGKHLSSM 158
    22-30] human PTHrP(1-30) ERVELLEKLLEK
    Haa(Laa Laa Haa Haa)2 Laa AVSEIQFMHNLGKHLSSM 159
    22-30] human PTHrP(1-30) RRRELLEKLLEK
  • Another useful PTH/PTHrP modulating domain has the sequence of the peptide known as TIP39: [0163]
    SLALADDAAFRERARLLAAL (SEQ ID NO: 160)
    ERRHWLNSYMHKLLVLDAP
  • TIP39 is described by Usdin et al. (1999), [0164] Nature Neurosci. 2(11): 941-3; Usdin et al. (1996), Endocrinology 137(10): 4285-97; Usdin et al. (1995), J. Biol. Chem. 270(26): 15455-8; Usdin et al. (1999), Endocrinol. 140(7): 3363-71.
  • Additional useful PTH/PTHrP modulating domain sequences may result from conservative and/or non-conservative modifications of the amino acid sequences of SEQ ID NOS: 3, 4, 5, TIP39, or the sequences listed in Tables 1 and 2. [0165]
  • Conservative modifications will produce peptides having functional and chemical characteristics similar to those of the PTH or PTHrP peptide from which such modifications are made. In contrast, substantial modifications in the functional and/or chemical characteristics of the peptides may be accomplished by selecting substitutions in the amino acid sequence that differ significantly in their effect on maintaining (a) the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the size of the molecule. [0166]
  • For example, a “conservative amino acid substitution” may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position. Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been previously described for “alanine scanning mutagenesis” (see, for example, MacLennan et al., 1998, [0167] Acta Physiol. Scand. Suppl. 643:55-67; Sasaki et al., 1998, Adv. Biophys. 35:1-24, which discuss alanine scanning mutagenesis).
  • Desired amino acid substitutions (whether conservative or non-conservative) can be determined by those skilled in the art at the time such substitutions are desired. For example, amino acid substitutions can be used to identify important residues of the peptide sequence, or to increase or decrease the affinity of the peptide or vehicle-peptide molecules (see preceding formulae) described herein. Exemplary amino acid substitutions are set forth in Table 3. [0168]
    TABLE 3
    Amino Acid Substitutions
    Original Exemplary Preferred
    Residues Substitutions Substitutions
    Ala Val, Leu, Ile Val
    Arg Lys, Gln, Asn Lys
    Asn Gln Gln
    Asp Glu Glu
    Cys Ser, Ala Ser
    Gln Asn Asn
    Glu Asp Asp
    Gly Pro, Ala Ala
    His Asn, Gln, Lys, Arg Arg
    Ile Leu, Val, Met, Ala, Leu
    Phe, Norleucine
    Leu Norleucine, Ile, Val, Ile
    Met, Ala, Phe
    Lys Arg, 1,4 Diamino- Arg
    butyric Acid, Gln, Asn
    Met Leu, Phe, Ile Leu
    Phe Leu, Val, Ile, Ala, Tyr Leu
    Pro Ala Gly
    Ser Thr, Ala, Cys Thr
    Thr Ser Ser
    Trp Tyr, Phe Tyr
    Tyr Trp, Phe, Thr, Ser Phe
    Val Ile, Met, Leu, Phe, Leu
    Ala, Norleucine
  • In certain embodiments, conservative amino acid substitutions also encompass non-naturally occurring amino acid residues which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. [0169]
  • As noted in the foregoing section “Definition of Terms,” naturally occurring residues may be divided into classes based on common sidechain properties that may be useful for modifications of sequence. For example, non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues may be introduced into regions of the peptide that are homologous with non-human orthologs, or into the non-homologous regions of the molecule. In addition, one may also make modifications using P or G for the purpose of influencing chain orientation. [0170]
  • In making such modifications, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5). [0171]
  • The importance of the hydropathic amino acid index in conferring interactive biological function on a protein is understood in the art. Kyte et al., [0172] J. Mol. Biol., 157: 105-131 (1982). It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making changes based upon the hydropathic index, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
  • It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. The greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein. [0173]
  • The following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (−0.4); proline (−0.5±1); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); tryptophan (−3.4). In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred. One may also identify epitopes from primary amino acid sequences on the basis of hydrophilicity. These regions are also referred to as “epitopic core regions.”[0174]
  • A skilled artisan will be able to determine suitable variants of the polypeptide as set forth in the foregoing sequences using well known techniques. For identifying suitable areas of the molecule that may be changed without destroying activity, one skilled in the art may target areas not believed to be important for activity. For example, when similar polypeptides with similar activities from the same species or from other species are known, one skilled in the art may compare the amino acid sequence of a peptide to similar peptides. With such a comparison, one can identify residues and portions of the molecules that are conserved among similar polypeptides. It will be appreciated that changes in areas of a peptide that are not conserved relative to such similar peptides would be less likely to adversely affect the biological activity and/or structure of the peptide. One skilled in the art would also know that, even in relatively conserved regions, one may substitute chemically similar amino acids for the naturally occurring residues while retaining activity (conservative amino acid residue substitutions). Therefore, even areas that may be important for biological activity or for structure may be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the peptide structure. [0175]
  • Additionally, one skilled in the art can review structure-function studies identifying residues in similar peptides that are important for activity or structure. In view of such a comparison, one can predict the importance of amino acid residues in a peptide that correspond to amino acid residues that are important for activity or structure in similar peptides. One skilled in the art may opt for chemically similar amino acid substitutions for such predicted important amino acid residues of the peptides. [0176]
  • One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar polypeptides. In view of that information, one skilled in the art may predict the alignment of amino acid residues of a peptide with respect to its three dimensional structure. One skilled in the art may choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art may generate test variants containing a single amino acid substitution at each desired amino acid residue. The variants can then be screened using activity assays know to those skilled in the art. Such data could be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed, undesirably reduced, or unsuitable activity, variants with such a change would be avoided. In other words, based on information gathered from such routine experiments, one skilled in the art can readily determine the amino acids where further substitutions should be avoided either alone or in combination with other mutations. [0177]
  • A number of scientific publications have been devoted to the prediction of secondary structure. See Moult J., [0178] Curr. Op. in Biotech., 7(4): 422-427 (1996), Chou et al., Biochemistry 13(2): 222-245 (1974); Chou et al., Biochemistry, 113(2): 211-222 (1974); Chou et al., Adv. Enzymol. Relat. Areas Mol. Biol., 47: 45-148 (1978); Chou et al., Ann. Rev. Biochem., 47: 251-276 and Chou et al., Biophys. J., 26: 367-384 (1979). Moreover, computer programs are currently available to assist with predicting secondary structure. One method of predicting secondary structure is based upon homology modeling. For example, two polypeptides or proteins which have a sequence identity of greater than 30%, or similarity greater than 40% often have similar structural topologies. The recent growth of the protein structural data base (PDB) has provided enhanced predictability of secondary structure, including the potential number of folds within a polypeptide's or protein's structure. See Holm et al., Nucl. Acid. Res., 27(1): 244-247 (1999). It has been suggested (Brenner et al., Curr. Op. Struct. Biol., 7(3): 369-376 (1997)) that there are a limited number of folds in a given polypeptide or protein and that once a critical number of structures have been resolved, structural prediction will gain dramatically in accuracy.
  • Additional methods of predicting secondary structure include “threading” (Jones, D., [0179] Curr. Opin. Struct. Biol., 7(3): 377-87 (1997); Sippl et al., Structure, 4(1): 15-9 (1996)), “profile analysis” (Bowie et al., Science, 253: 164-170 (1991); Gribskov et al., Meth. Enzym., 183: 146-159 (1990); Gribskov et al., Proc. Nat. Acad. Sci., 84(13): 4355-8 (1987)), and “evolutionary linkage” (See Home, supra, and Brenner, supra).
  • Vehicles. This invention requires the presence of at least one vehicle (F[0180] 1) attached to a peptide through the C-terminus or a sidechain of one of the amino acid residues. Multiple vehicles may also be used; e.g., an Fc at the C-terminus and a PEG group at a sidechain.
  • An Fc domain is the preferred vehicle. The Fc domain may be fused to the C terminus of the peptides. [0181]
  • As noted above, Fc variants are suitable vehicles within the scope of this invention. A native Fc may be extensively modified to form an Fc variant in accordance with this invention, provided binding to the salvage receptor is maintained; see, for example WO 97/34631 and WO 96/32478. In such Fc variants, one may remove one or more sites of a native Fc that provide structural features or functional activity not required by the fusion molecules of this invention. One may remove these sites by, for example, substituting or deleting residues, inserting residues into the site, or truncating portions containing the site. The inserted or substituted residues may also be altered amino acids, such as peptidomimetics or D-amino acids. Fc variants may be desirable for a number of reasons, several of which are described below. Exemplary Fc variants include molecules and sequences in which: [0182]
  • 1. Sites involved in disulfide bond formation are removed. Such removal may avoid reaction with other cysteine-containing proteins present in the host cell used to produce the molecules of the invention. For this purpose, the cysteine-containing segment at the N-terminus may be truncated or cysteine residues may be deleted or substituted with other amino acids (e.g., alanyl, seryl). In particular, one may truncate the N-terminal 20-amino acid segment of SEQ ID NO: 2 or delete or substitute the cysteine residues at [0183] positions 7 and 10 of SEQ ID NO: 2. Even when cysteine residues are removed, the single chain Fc domains can still form a dimeric Fc domain that is held together non-covalently.
  • 2. A native Fc is modified to make it more compatible with a selected host cell. For example, one may remove the PA sequence near the N-terminus of a typical native Fc, which may be recognized by a digestive enzyme in [0184] E. coli such as proline iminopeptidase. One may also add an N-terminal methionine residue, especially when the molecule is expressed recombinantly in a bacterial cell such as E. coli. The Fc domain of SEQ ID NO: 2 is one such Fc variant.
  • 3. A portion of the N-terminus of a native Fc is removed to prevent N-terminal heterogeneity when expressed in a selected host cell. For this purpose, one may delete any of the first 20 amino acid residues at the N-terminus, particularly those at [0185] positions 1, 2, 3, 4 and 5.
  • 4. One or more glycosylation sites are removed. Residues that are typically glycosylated (e.g., asparagine) may confer cytolytic response. Such residues may be deleted or substituted with unglycosylated residues (e.g., alanine). [0186]
  • 5. Sites involved in interaction with complement, such as the C1q binding site, are removed. For example, one may delete or substitute the EKK sequence of human IgG1. Complement recruitment may not be advantageous for the molecules of this invention and so may be avoided with such an Fc variant. [0187]
  • 6. Sites are removed that affect binding to Fc receptors other than a salvage receptor. A native Fc may have sites for interaction with certain white blood cells that are not required for the fusion molecules of the present invention and so may be removed. [0188]
  • 7. The ADCC site is removed. ADCC sites are known in the art; see, for example, [0189] Molec. Immunol. 29 (5): 633-9 (1992) with regard to ADCC sites in IgG1. These sites, as well, are not required for the fusion molecules of the present invention and so may be removed.
  • 8. When the native Fc is derived from a non-human antibody, the native Fc may be humanized. Typically, to humanize a native Fc, one will substitute selected residues in the non-human native Fc with residues that are normally found in human native Fc. Techniques for antibody humanization are well known in the art. [0190]
  • Preferred Fc variants include the following. In SEQ ID NO: 2 (FIG. 4) the leucine at [0191] position 15 may be substituted with glutamate; the glutamate at position 99, with alanine; and the lysines at positions 101 and 103, with alanines. In addition, one or more tyrosine residues can be replaced by phenyalanine residues.
  • An alternative vehicle would be a protein, polypeptide, peptide, antibody, antibody fragment, or small molecule (e.g., a peptidomimetic compound) capable of binding to a salvage receptor. For example, one could use as a vehicle a polypeptide as described in U.S. Pat. No. 5,739,277, issued Apr. 14, 1998 to Presta et al. Peptides could also be selected by phage display or RNA-peptide screening for binding to the FcRn salvage receptor. Such salvage receptor-binding compounds are also included within the meaning of “vehicle” and are within the scope of this invention. Such vehicles should be selected for increased half-life (e.g., by avoiding sequences recognized by proteases) and decreased immunogenicity (e.g., by favoring non-immunogenic sequences, as discovered in antibody humanization). [0192]
  • As noted above, polymer vehicles may also be used for F[0193] 1 and F2. Various means for attaching chemical moieties useful as vehicles are currently available, see, e.g., Patent Cooperation Treaty (“PCT”) International Publication No. WO 96/11953, entitled “N-Terminally Chemically Modified Protein Compositions and Methods,” herein incorporated by reference in its entirety. This PCT publication discloses, among other things, the selective attachment of water soluble polymers to the N-terminus of proteins.
  • A preferred polymer vehicle is polyethylene glycol (PEG). The PEG group may be of any convenient molecular weight and may be linear or branched. The average molecular weight of the PEG will preferably range from about 2 kiloDalton (“kD”) to about 100 kD, more preferably from about 5 kD to about 50 kD, most preferably from about 5 kD to about 10 kD. The PEG groups will generally be attached to the compounds of the invention via acylation or reductive alkylation through a reactive group on the PEG moiety (e.g., an aldehyde, amino, thiol, or ester group) to a reactive group on the inventive compound (e.g., an aldehyde, amino, or ester group). [0194]
  • A useful strategy for the PEGylation of synthetic peptides consists of combining, through forming a conjugate linkage in solution, a peptide and a PEG moiety, each bearing a special functionality that is mutually reactive toward the other. The peptides can be easily prepared with conventional solid phase synthesis. The peptides are “preactivated” with an appropriate functional group at a specific site. The precursors are purified and fully characterized prior to reacting with the PEG moiety. Ligation of the peptide with PEG usually takes place in aqueous phase and can be easily monitored by reverse phase analytical HPLC. The PEGylated peptides can be easily purified by preparative HPLC and characterized by analytical HPLC, amino acid analysis and laser desorption mass spectrometry. [0195]
  • Polysaccharide polymers are another type of water soluble polymer which may be used for protein modification. Dextrans are polysaccharide polymers comprised of individual subunits of glucose predominantly linked by α1-6 linkages. The dextran itself is available in many molecular 25 weight ranges, and is readily available in molecular weights from about 1 kD to about 70 kD. Dextran is a suitable water soluble polymer for use in the present invention as a vehicle by itself or in combination with another vehicle (e.g., Fc). See, for example, WO 96/11953 and WO 96/05309. The use of dextran conjugated to therapeutic or diagnostic immunoglobulins has been reported; see, for example, European Patent Publication No. 0 315 456, which is hereby incorporated by reference in its entirety. Dextran of about 1 kD to about 20 kD is preferred when dextran is used as a vehicle in accordance with the present invention. [0196]
  • Linkers. Any “linker” group is optional. When present, its chemical structure is not critical, since it serves primarily as a spacer. The linker is preferably made up of amino acids linked together by peptide bonds. Thus, in preferred embodiments, the linker is made up of from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids. Some of these amino acids may be glycosylated, as is well understood by those in the art. In a more preferred embodiment, the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine. Even more preferably, a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine. Thus, preferred linkers are polyglycines (particularly (Gly)[0197] 4, (Gly)5), poly(Gly-Ala), and polyalanines. Other specific examples of linkers are:
    (Gly)3Lys(Gly)4; (SEQ ID NO: 6)
    (Gly)3AsnGlySer(Gly)2; (SEQ ID NO: 7)
    (Gly)3Cys(Gly)4; and (SEQ ID NO: 8)
    GlyProAsnGlyGly. (SEQ ID NO: 9)
  • To explain the above nomenclature, for example, (Gly)[0198] 3Lys(Gly)4 means Gly-Gly-Gly-Lys-Gly-Gly-Gly-Gly. Combinations of Gly and Ala are also preferred. The linkers shown here are exemplary; linkers within the scope of this invention may be much longer and may include other residues.
  • Non-peptide linkers are also possible. For example, alkyl linkers such as —NH—(CH[0199] 2)5—C(O)—, wherein s=2-20 could be used. These alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., C1-C6) lower acyl, halogen (e.g., Cl, Br), CN, NH2, phenyl, etc. An exemplary non-peptide linker is a PEG linker, VI
    Figure US20040214996A1-20041028-C00001
  • wherein n is such that the linker has a molecular weight of 100 to 5000 kD, preferably 100 to 500 kD. The peptide linkers may be altered to form derivatives in the same manner as described above. [0200]
  • Derivatives. The inventors also contemplate derivatizing the peptide and/or vehicle portion of the compounds. Such derivatives may improve the solubility, absorption, biological half life, and the like of the compounds. The moieties may alternatively eliminate or attenuate any undesirable side-effect of the compounds and the like. Exemplary derivatives include compounds in which: [0201]
  • 1. The compound or some portion thereof is cyclic. For example, the peptide portion may be modified to contain two or more Cys residues (e.g., in the linker), which could cyclize by disulfide bond formation. [0202]
  • 2. The compound is cross-linked or is rendered capable of cross-linking between molecules. For example, the peptide portion may be modified 20 to contain one Cys residue and thereby be able to form an intermolecular disulfide bond with a like molecule. The compound may also be cross-linked through its C-terminus, as in the molecule shown below. V [0203]
    Figure US20040214996A1-20041028-C00002
  • 3. One or more peptidyl [—C(O)NR—] linkages (bonds) is replaced by a non-peptidyl linkage. Exemplary non-peptidyl linkages are —CH[0204] 2-carbamate [—CH2—OC(O)NR—], phosphonate, —CH2-sulfonamide [—CH2—S(O)2NR—], urea [—NHC(O)NH—], —CH2-secondary amine, and alkylated peptide [—C(O)NR6— wherein R6 is lower alkyl].
  • 4. The N-terminus is derivatized. Typically, the N-terminus may be acylated or modified to a substituted amine. Exemplary N-terminal derivative groups include —NRR[0205] 1 (other than —NH2), —NRC(O)R1, —NRC(O)OR1, —NRS(O)2R1, —NHC(O)NHR1, succinimide, or benzyloxycarbonyl-NH— (CBZ-NH—), wherein R and R1 are each independently hydrogen or lower alkyl and wherein the phenyl ring may be substituted with 1 to 3 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, chloro, and bromo.
  • 5. The free C-terminus is derivatized. Typically, the C-terminus is esterified or amidated. Exemplary C-terminal derivative groups include, for example, —C(O)R[0206] 2 wherein R2 is lower alkoxy or —NR3R4 wherein R3 and R4 are independently hydrogen or C1-C8 alkyl (preferably C1-C4 alkyl).
  • 6. A disulfide bond is replaced with another, preferably more stable, cross-linking moiety (e.g., an alkylene). See, e.g., Bhatnagar et al. (1996), [0207] J. Med. Chem. 39: 3814-9; Alberts et al. (1993) Thirteenth Am. Pep. Symp., 357-9.
  • 7. One or more individual amino acid residues is modified. Various derivatizing agents are known to react specifically with selected sidechains or terminal residues, as described in detail below. [0208]
  • Lysinyl residues and amino terminal residues may be reacted with succinic or other carboxylic acid anhydrides, which reverse the charge of the lysinyl residues. Other suitable reagents for derivatizing alpha-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate. [0209]
  • Arginyl residues may be modified by reaction with any one or combination of several conventional reagents, including phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginyl residues requires that the reaction be performed in alkaline conditions because of the high pKa of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group. [0210]
  • Specific modification of tyrosyl residues has been studied extensively, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizole and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively. [0211]
  • Carboxyl sidechain groups (aspartyl or glutamyl) may be selectively modified by reaction with carbodiimides (R′—N═C═N—R′) such as 1-cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl) carbodiimide. Furthermore, aspartyl and glutamyl residues may be converted to asparaginyl and glutaminyl residues by reaction with ammonium ions. [0212]
  • Glutaminyl and asparaginyl residues may be deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention. [0213]
  • Cysteinyl residues can be replaced by amino acid residues or other moieties either to eliminate disulfide bonding or, conversely, to stabilize cross-linking. See, e.g., Bhatnagar et al. (1996), [0214] J. Med. Chem. 39: 3814-9.
  • Derivatization with bifunctional agents is useful for cross-linking the peptides or their functional derivatives to a water-insoluble support matrix or to other macromolecular vehicles. Commonly used cross-linking agents include, e.g., 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3′-dithiobis(succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane. Derivatizing agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light. Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization. [0215]
  • Carbohydrate (oligosaccharide) groups may conveniently be attached to sites that are known to be glycosylation sites in proteins. Generally, O-linked oligosaccharides are attached to serine (Ser) or threonine (Thr) residues while N-linked oligosaccharides are attached to asparagine (Asn) residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline. X is preferably one of the 19 naturally occurring amino acids other than proline. The structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different. One type of sugar that is commonly found on both is N-acetylneuraminic acid (referred to as sialic acid). Sialic acid is usually the terminal residue of both N-linked and O-linked oligosaccharides and, by virtue of its negative charge, may confer acidic properties to the glycosylated compound. Such site(s) may be incorporated in the linker of the compounds of this invention and are preferably glycosylated by a cell during recombinant production of the polypeptide compounds (e.g., in mammalian cells such as CHO, BHK, COS). However, such sites may further be glycosylated by synthetic or semi-synthetic procedures known in the art. [0216]
  • Other possible modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, oxidation of the sulfur atom in Cys, methylation of the alpha-amino groups of lysine, arginine, and histidine side chains. Creighton, [0217] Proteins: Structure and Molecule Properties (W. H. Freeman & Co., San Francisco), pp. 79-86 (1983).
  • Compounds of the present invention may be changed at the DNA level, as well. The DNA sequence of any portion of the compound may be changed to codons more compatible with the chosen host cell. For [0218] E. coli, which is the preferred host cell, optimized codons are known in the art. Codons may be substituted to eliminate restriction sites or to include silent restriction sites, which may aid in processing of the DNA in the selected host cell. The vehicle, linker and peptide DNA sequences may be modified to include any of the foregoing sequence changes.
  • Methods of Making [0219]
  • The compounds of this invention largely may be made in transformed host cells using recombinant DNA techniques. To do so, a recombinant DNA molecule coding for the peptide is prepared. Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidate method. Also, a combination of these techniques could be used. [0220]
  • The invention also includes a vector capable of expressing the peptides in an appropriate host. The vector comprises the DNA molecule that codes for the peptides operatively linked to appropriate expression control sequences. Methods of effecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation. [0221]
  • The resulting vector having the DNA molecule thereon is used to transform an appropriate host. This transformation may be performed using methods well known in the art. [0222]
  • Any of a large number of available and well-known host cells may be used in the practice of this invention. The selection of a particular host is dependent upon a number of factors recognized by the art. These include, for example, compatibility with the chosen expression vector, toxicity of the peptides encoded by the DNA molecule, rate of transformation, ease of recovery of the peptides, expression characteristics, bio-safety and costs. A balance of these factors must be struck with the understanding that not all hosts may be equally effective for the expression of a particular DNA sequence. Within these general guidelines, useful microbial hosts include bacteria (such as [0223] E. coli sp.), yeast (such as Saccharomyces sp.) and other fungi, insects, plants, mammalian (including human) cells in culture, or other hosts known in the art.
  • Next, the transformed host is cultured and purified. Host cells may be cultured under conventional fermentation conditions so that the desired compounds are expressed. Such fermentation conditions are well known in the art. Finally, the peptides are purified from culture by methods well known in the art. [0224]
  • The compounds may also be made by synthetic methods. For example, solid phase synthesis techniques may be used. Suitable techniques are well known in the art, and include those described in Merrifield (1973), [0225] Chem. Polypeptides, pp. 335-61 (Katsoyannis and Panayotis eds.); Merrifield (1963), J. Am. Chem. Soc. 85: 2149; Davis et al. (1985), Biochem. Intl. 10: 394-414; Stewart and Young (1969), Solid Phase Peptide Synthesis; U.S. Pat. No. 3,941,763; Finn et al. (1976), The Proteins (3rd ed.) 2: 105-253; and Erickson et al. (1976), The Proteins (3rd ed.) 2: 257-527. Solid phase synthesis is the preferred technique of making individual peptides since it is the most cost-effective method of making small peptides.
  • Compounds that contain derivatized peptides or which contain non-peptide groups may be synthesized by well-known organic chemistry techniques. [0226]
  • Uses of the Compounds [0227]
  • The compounds of this invention have pharmacologic activity resulting from their interaction with PTH-1 receptor or PTH-2 receptor. Mannstadt et al. (1999), [0228] Am. J. Physiol. 277. 5Pt 2. F665-75. PTH and agonists thereof increase bone resorption, increase renal calcium reabsorption, decrease epidermal proliferation, and decrease hair growth. Holick et al. (1994) Proc. Natl. Sci. USA 91 (17): 8014-6; Schilli et al. (1997), J. Invest. Dermatol. 108(6): 928-32. Thus, antagonists of PTH-1 receptor and/or PTH-2 receptor are useful in treating:
  • primary and secondary hyperparathyroidism; [0229]
  • hypercalcemia, including hypercalcemia resulting from solid tumors (breast, lung and kidney) and hematologic malignacies (multiple myeloma, lymphoma and leukemia); idiopathic hypercalcemia, and hypercalcemia associated with hyperthyroidism and renal function disorders; [0230]
  • tumor metastases, particularly metastases to bone, and particularly related to breast and prostate cancer; [0231]
  • cachexia and anorexia, particularly as associated with cancer; [0232]
  • osteopenia that is related to or aggravated by aberrant PTH receptor signaling, including various forms of osteoporosis, such as: [0233]
  • primary osteoporosis; [0234]
  • post-menopausal and age-related osteoporosis; [0235]
  • endocrine osteoporosis (hyperthyroidism, hyperparathyroidism, Cushing's syndrome, and acromegaly); [0236]
  • hereditary and congenital forms of osteoporosis (e.g., osteogenesis imperfecta, homocystinuria, Menkes' syndrome, and Riley-Day syndrome); [0237]
  • osteoporosis due to immobilization of extremities; [0238]
  • osteoporosis secondary to other disorders, such as hemochromatosis, hyperprolactinemia, anorexia nervosa, thyrotoxicosis, diabetes mellitus, celiac disease, inflammatory bowel disease, primary biliary cirrhosis, rheumatoid arthritis, ankylosing spondylitis, multiple myeloma, lymphoproliferative diseases, and systemic mastocytosis; [0239]
  • osteoporosis secondary to surgery (e.g., gastrectomy) or to drug therapy, such as chemotherapy, anticonvulsant therapy, immunosuppressive therapy, and anticoagulant therapy; [0240]
  • osteoporosis secondary to glucocorticosteroid treatment for such diseases as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), asthma, temporal arteritis, vasculitis, chronic obstructive pulmonary disease, polymyalgia rheumatica, polymyositis, chronic interstitial lung disease; [0241]
  • osteoporosis secondary to glucocorticosteroid and/or immunomodulatory treatment to prevent organ rejection following organ transplant such as kidney, liver, lung, heart transplants; [0242]
  • osteoporosis due to submission to microgravity, such as observed during space travel; [0243]
  • osteoporosis associated with malignant disease, such as breast cancer, prostate cancer; [0244]
  • Paget's disease of bone (osteitis deformans) in adults and juveniles; [0245]
  • osteomyelitis, or an infectious lesion in bone, leading to bone loss; [0246]
  • osteopenia following surgery, induced by steroid administration, and associated with disorders of the small and large intestine and with chronic hepatic and renal diseases. [0247]
  • Osteonecrosis, or bone cell death, associated with traumatic injury or nontraumatic necrosis associated with Gaucher's disease, sickle cell anemia, systemic lupus erythematosus, rheumatoid arthritis, periodontal disease, osteolytic metastasis, and other conditions; [0248]
  • alopecia (deficient hair growth or partial or complete hair loss), including androgenic alopecia (male pattern baldness), toxic alopecia, alopecia senilis, alopecia areata, alopecia pelada, and trichotillomania; [0249]
  • and the like. [0250]
  • There are other conditions wherein a patient would benefit from the activity of PTH or PTHrP. For those indications, PTH receptor agonists are useful as a therapeutic treatment. In particular, such indications include fracture repair (including healing of non-union fractures), osteopenia, including various forms of osteoporosis, such as: [0251]
  • primary osteoporosis; [0252]
  • post-menopausal and age-related osteoporosis; [0253]
  • endocrine osteoporosis (hyperthyroidism, Cushing's syndrome, and acromegaly); [0254]
  • hereditary and congenital forms of osteoporosis (e.g., osteogenesis imperfecta, homocystinuria, Menkes' syndrome, and Riley-Day syndrome); [0255]
  • osteoporosis due to immobilization of extremities; [0256]
  • osteoporosis secondary to other disorders, such as hemochromatosis, hyperprolactinemia, anorexia nervosa, thyrotoxicosis, diabetes mellitus, celiac disease, inflammatory bowel disease, primary biliary cirrhosis, rheumatoid arthritis, ankylosing spondylitis, multiple myeloma, lymphoproliferative diseases, and systemic mastocytosis; [0257]
  • osteoporosis secondary to surgery (e.g., gastrectomy) or to drug therapy, such as chemotherapy, anticonvulsant therapy, immunosuppressive therapy, and anticoagulant therapy; [0258]
  • osteoporosis secondary to glucocorticosteroid treatment for diseases such as RA, SLE, asthma, temporal arteritis, vasculitis, chronic obstructive pulmonary disease, polymyalgia rheumatica, polymyositis, chronic interstitial lung disease; [0259]
  • osteoporosis secondary to glucocorticosteroid and/or immunomodulatory treatment to prevent organ rejection following organ transplant such as kidney, liver, lung, heart transplants; [0260]
  • osteoporosis due to submission to microgravity, such as observed during space travel; [0261]
  • osteoporosis associated with malignant disease, such as breast cancer, prostate cancer; [0262]
  • PTH agonists with extended half-life (e.g., those linked to Fc domains) may be used with an inhibitor of bone resorption. Inhibitors of bone resorption include OPG and OPG derivatives, OPG-L (RANKL) antibody, calcitonin (e.g., Miacalcin®, Calcimar®), bisphosphonates (e.g., APD, alendronate, risedronate, etidronate, pamidronate, tiludronate, clodronate, neridronate, ibandronate, zoledronate), estrogens (e.g., Premarin®, Estraderm®, Prempro®, Alora®, Climara®, Vivelle®, Estratab® Ogen®), selective estrogen receptor modulators (e.g., raloxifene, droloxifene, lasofoxifene), tibolone, and the like. Exemplary bone resorption inhibitors are described in WO98/46751 and WO97/23614, which are hereby incorporated by reference in their entirety. [0263]
  • The compounds of this invention may be appropriate as a monotherapy for the treatment of Osteoporosis, and it is possible that the addition of an antiresorptive agent to PTH-Fc treatment will increase both their efficacy and therapeutic window. Both PTH and PTH-Fc cause an increase in both bone formation and bone resorption. The ability of antiresorptives to block the osteoclast response could limit the hypercalcemic effects of PTH-Fc and could also increase bone mas [0264]
  • Pharmaceutical Compositions [0265]
  • In General. The present invention also provides methods of using pharmaceutical compositions of the inventive compounds. Such pharmaceutical compositions may be for administration for injection, or for oral, pulmonary, nasal, transdermal or other forms of administration. In general, the invention encompasses pharmaceutical compositions comprising effective amounts of a compound of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents of various buffer content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., [0266] Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc. or into liposomes. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation. 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 in their entirety. The compositions may be prepared in liquid form, or may be in dried powder, such as lyophilized form. Implantable sustained release formulations are also contemplated, as are transdermal formulations.
  • Twice weekly dosing of the compounds of this invention is superior to daily injection of PTH (1-34) for increasing osteoblast number, bone volume, and bone mineral density in rodents. In adult mice, twice weekly dosing with PTH-(1-34)-Fc caused greater increases in bone density and bone volume compared to daily PTH-(1-34). (See FIG. 10.) In an aged OVX rat model of osteoporosis, twice weekly dosing was able to reverse more than 50% of the bone loss induced by one year of estrogen ablation. The effect seen in the aged rat model was even greater when combined with a bisphosphonate (APD). In rats, a single SC injection of PTH-(1-34)-Fc (340 nmol/kg) caused a hypercalcemic response which persisted for 72 hours (FIG. 8). This duration is concordant with the rate of clearance of PTH-(1-34)-Fc from the serum, and is consistent with an optimal twice-weekly dosing regimen in rats. [0267]
  • The optimal dosing of primates may be less frequent compared to rats or mice. Weekly (or less frequent) dosing may be optimal in primates, based on the observation that the hypercalcemic response of OVX cynomolgus monkeys to a single subcutaneous injection of PTH-(1-34)-Fc (10-34 nmol/kg) persisted for about 168 hours (FIG. 11). This observation suggests that a single subcutaneous dose of PTH-(1-34)-Fc in primates is cleared within about 1 week, which could also represent the maximum dosing frequency required for anabolic effects. [0268]
  • Oral dosage forms. Contemplated for use herein are oral solid dosage forms, which are described generally in Chapter 89 of [0269] Remington's Pharmaceutical Sciences (1990), 18th Ed., Mack Publishing Co. Easton Pa. 18042, which is herein incorporated by reference in its entirety. Solid dosage forms include tablets, capsules, pills, troches or lozenges, cachets or pellets. Also, liposomal or proteinoid encapsulation may be used to formulate the present compositions (as, for example, proteinoid microspheres reported in U.S. Pat. No. 4,925,673). Liposomal encapsulation may be used and the liposomes may be derivatized with various polymers (e.g., U.S. Pat. No. 5,013,556). A description of possible solid dosage forms for the therapeutic is given in Chapter 10 of Marshall, K., Modern Pharmaceutics (1979), edited by G. S. Banker and C. T. Rhodes, herein incorporated by reference in its entirety. In general, the formulation will include the inventive compound, and inert ingredients which allow for protection against the stomach environment, and release of the biologically active material in the intestine.
  • Also specifically contemplated are oral dosage forms of the above inventive compounds. If necessary, the compounds may be chemically modified so that oral delivery is efficacious. Generally, the chemical modification contemplated is the attachment of at least one moiety to the compound 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 compound and increase in circulation time in the body. Moieties useful as covalently attached vehicles in this invention may also be used for this purpose. Examples of such moieties include: PEG, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. See, for example, Abuchowski and Davis, [0270] Soluble Polymer-Enzyme Adducts Enzymes as Drugs (1981), Hocenberg and Roberts, eds., Wiley-Interscience, New York, N.Y., pp. 367-83; Newmark, et al. (1982), J. Appl. Biochem. 4:185-9. Other polymers that could be used are poly-1,3-dioxolane and poly-1,3,6-tioxocane. Preferred for pharmaceutical usage, as indicated above, are PEG moieties.
  • For oral delivery dosage forms, it is also possible to use a salt of a modified aliphatic amino acid, such as sodium N-(8-[2-hydroxybenzoyl]amino) caprylate (SNAC), as a carrier to enhance absorption of the therapeutic compounds of this invention. The clinical efficacy of a heparin formulation using SNAC has been demonstrated in a Phase II trial conducted by Emisphere Technologies. See U.S. Pat. No. 5,792,451, “Oral drug delivery composition and methods”. [0271]
  • The compounds of this invention can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets. The therapeutic could be prepared by compression. [0272]
  • Colorants and flavoring agents may all be included. For example, the protein (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. [0273]
  • One may dilute or increase the volume of the compound of the invention 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 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. [0274]
  • Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrants include but are not limited to starch including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used. Another form of the disintegrants are the insoluble cationic exchange resins. Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants. [0275]
  • Binders may be used to hold the therapeutic 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 hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic. [0276]
  • An antifrictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic 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. [0277]
  • Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate. [0278]
  • To aid dissolution of the compound of this invention 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 benzalkonium chloride or benzethonium chloride. The list of potential nonionic detergents that could be included in the formulation as surfactants are lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated [0279] castor oil 10, 50 and 60, glycerol monostearate, polysorbate 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 may also be included in the formulation to enhance uptake of the compound. Additives potentially having this property are for instance the fatty acids oleic acid, linoleic acid and linolenic acid. [0280]
  • Controlled release formulation may be desirable. The compound of this invention 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, e.g., alginates, polysaccharides. Another form of a controlled release of the compounds of this invention 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. Some enteric coatings also have a delayed release effect. [0281]
  • Other coatings may be used for the formulation. These include a variety of sugars which could be applied in a coating pan. The therapeutic agent 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 nonenteric 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 phthalic acid. [0282]
  • 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. [0283]
  • Pulmonary delivery forms. Also contemplated herein is pulmonary delivery of the present protein (or derivatives thereof). The protein (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream. (Other reports of this include Adjei et al., [0284] Pharma. Res. (1990) 7: 565-9; Adjei et al. (1990), Internatl. J. Pharmaceutics 63: 135-44 (leuprolide acetate); Braquet et al. (1989), J. Cardiovasc. Pharmacol. 13 (suppl.5): s.143-146 (endothelin-1); Hubbard et al. (1989), Annals Int. Med. 3: 206-12 (α1-antitrypsin); Smith et al. (1989), J. Clin. Invest. 84: 1145-6 (α1-proteinase); Oswein et al. (March 1990), “Aerosolization of Proteins”, Proc. Symp. Resp. Drug Delivery II, Keystone, Colorado (recombinant human growth hormone); Debs et al. (1988), J. Immunol. 140: 3482-8 (interferon-γ and tumor necrosis factor α) and Platz et al., U.S. Pat. No. 5,284,656 (granulocyte colony stimulating factor).
  • 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, manufactured by Mallinckrodt, Inc., St. Louis, Mo.; the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colo.; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, N.C.; and the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Mass. [0285]
  • All such devices require the use of formulations suitable for the dispensing of the inventive compound. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to diluents, adjuvants and/or carriers useful in therapy. [0286]
  • The inventive compound should most advantageously be prepared in particulate form with an average particle size of less than 10 μm (or microns), most preferably 0.5 to 5 μm, for most effective delivery to the distal lung. [0287]
  • Pharmaceutically acceptable carriers include carbohydrates such as trehalose, mannitol, xylitol, sucrose, lactose, and sorbitol. Other ingredients for use in formulations may include DPPC, DOPE, DSPC and DOPC. Natural or synthetic surfactants may be used. PEG may be used (even apart from its use in derivatizing the protein or analog). Dextrans, such as cyclodextran, may be used. Bile salts and other related enhancers may be used. Cellulose and cellulose derivatives may be used. Amino acids may be used, such as use in a buffer formulation. [0288]
  • Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated. [0289]
  • Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise the inventive compound 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 protein caused by atomization of the solution in forming the aerosol. [0290]
  • Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the inventive compound 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 trichlorofluoromethane, 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. [0291]
  • Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing the inventive compound and may also include a bulking agent, such as lactose, sorbitol, sucrose, mannitol, trehalose, or xylitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation. [0292]
  • Nasal delivery forms. Nasal delivery of the inventive compound is also contemplated. Nasal delivery allows the passage of the protein 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. Delivery via transport across other mucous membranes is also contemplated. [0293]
  • Dosages. The dosage regimen involved in a method for treating the above-described conditions will be determined by the attending physician, considering various factors which modify the action of drugs, e.g. the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. Generally, the daily regimen should be in the range of 0.1-1000 micrograms of the inventive compound per kilogram of body weight, preferably 0.1-150 micrograms per kilogram. [0294]
  • Specific Preferred Embodiments [0295]
  • The inventors have determined preferred structures for the preferred peptides listed in Table 4 below. The symbol “A” may be any of the linkers described herein or may simply represent a normal peptide bond (i.e., so that no linker is present). Tandem repeats and linkers are shown separated by dashes for clarity. [0296]
    TABLE 4
    Preferred embodiments
    Peptide SEQ
    Sequence/structure description ID NO:
    SVSEIQLMHNLGKHLNSMERVEWL PTH(1-34) 161
    RKKLQDVHNF-Λ-F1
    SVSEIQLMHNRGKHLNSMERVEWL (L11R) 162
    RKKLQDVHNF-Λ-F1 PTH(1-34)
    SVSEIQLMHNKGKHLNSMERVEWL (L11K) 163
    RKKLQDVHNF-Λ-F1 PTH(1-34)
    SVSEIQLMHNLGKHLNSMRRVEWL (E19R) 164
    RKKLQDVHNF-Λ-F1 PTH(1-34)
    SVSEIQLMHNLGKHLNSMERVEWL PTH(1-31) 165
    RKKLQDV-Λ-F1
    SVSEIQLMHNLGKHLNSMERVEWL PTH(1-30) 166
    RKKLQD-Λ-F1
    F1-Λ-SVSEIQLMHNLGKHLNSME PTH(1-29) 167
    RVEWLRKKLQ
    F1-Λ-SVSEIQLMHNLGKHLNSME PTH(1-28) 168
    RVEWLRKKL
    LLHNLGKSIQDLRRRFFLHHLIAE (D10N, K11L) 169
    IHTA-Λ-F1 PTHrP(7-34)
    SLALADDAAFRERARLLAALERRH TIP39 170
    WLNSYMHKLLVLDAP-Λ-F1
  • “F[0297] 1” is an Fc domain as defined previously herein. In addition to those listed in Table 4, the inventors further contemplate heterodimers in which each strand of an Fc dimer is linked to a different peptide sequence; for example, a molecule in which one strand can be described by SEQ ID NO: 166, the other by SEQ ID NO: 170 or an Fc linked with any of the sequences in Tables 1 and 2.
  • All of the compounds of this invention can be prepared by methods described in PCT appl. no. WO 99/25044. [0298]
  • The invention will now be further described by the following working examples, which are illustrative rather than limiting. [0299]
  • EXAMPLE 1 Bioactivity of an Fc-conjugated PTH/PTHrP Receptor (PTH-R1) Agonist [PTH-(1-34)-Fc]
  • Introduction [0300]
  • Parathyroid hormone [PTH-(1-34) or native PTH-(1-84)] causes increased bone formation and increased bone mass when injected daily. This anabolic response was previously thought to require brief exposure to PTH, which is facilitated by the short half-life (less than 1 h) of PTH. Clinically, the anabolic effect of PTH therapy requires daily SC injection, which is a significant barrier to the widespread use of PTH. Less frequent injections of PTH would be clinically desirable and could be achieved by increasing the in vivo half-life of PTH. Short-term (intermittent) exposure to PTH (<1 h/day) stimulates osteoblastic bone formation, while long-term (continuous) exposure (>2 h/day) stimulates osteoclastic bone resorption (Dobnig et al, [0301] Endocrinology 138: 4607, 1998). The art suggests that PTH with an extended half-life on its own may increase bone resorption and lead to hypercalcemia. However, it should be possible to prevent PTH-induced osteoclast activity with bone resorption inhibitors. Osteoprotegerin (OPG) may be well suited for this purpose. A single treatment of rats, mice or humans with OPG-Fc causes sustained inhibition of bone resorption, by essentially eradicating the osteoclast population. Co-administration of a potent bone resorption inhibitor, like OPG, may provide greater effect. This regimen would theoretically permit the unopposed stimulation of bone formation by PTH, leading to increased bone mass. It is likely that other bone resorption inhibitors, including bisphosphonates or estrogen, would also inhibit PTH-induced bone resorption and could therefore be used in combination with a long-acting PTH molecule. Towards this goal, we have cloned, expressed and purified human PTH-(1-34)-Fc. Fc conjugation of proteins causes a significant increase in their circulating half life, which may permit injections of PTH-(1-34)-Fc on a schedule similar to or identical to that of OPG-Fc. The benefits of this invention include less frequent injections of PTH, from the current standard of once per day to as infrequently as once per quarter.
  • Materials, Methods, and Results [0302]
  • Hypercalcemia Assay [0303]
  • We tested the potency and duration of effect of PTH-(1-34)-Fc in a murine hypercalcemia model. Briefly, mice were injected once SC with varying doses of PTH-(1-34) or PTH-(1-34)-Fc, and peripheral blood was collected from the retroorbital sinus for determination of blood ionized calcium. The half-life and the potency of PTH-(1-34)-Fc was greater than that of PTH-(1-34), as evidenced by the sustained hypercalcemic response of mice to the former agent (FIG. 4). Hypercalcemia induced by PTH-(1-34) persisted for 6-24 h, while equimolar doses of PTH-(1-34)-Fc caused more sustained hypercalcemia (48-72 h). This duration of response is consistent with greater half-life of the PTH-(1-34)-Fc construct vs. PTH-(1-34). The potency of PTH-(1-34)-Fc was also significantly greater than that of PTH-(1-34) (FIG. 4). The highest dose of PTH-(1-34)-Fc caused a greater increase in peak ionized calcium levels compared with an equimolar dose of PTH-(1-34). Analysis of the area under the curve (AUC) demonstrated that at the highest dose employed, PTH-(1-34)-Fc caused a 2.6-fold greater hypercalcemic response than did equimolar doses of PTH-(1-34). [0304]
  • Anabolic Assay [0305]
  • Having demonstrated the superior pharmacology and half-life of PTH-(1-34)-Fc over PTH-(1-34), we conducted a pilot study to determine whether PTH-(1-34)-Fc co-treatment with OPG-Fc would increase bone mass. Briefly, 6-month-old male Sprague Dawley (SD) rats were divided into groups of 6. Baseline bone mineral density (BMD) was determined in the third lumbar vertebra (L3) of all rats by dual-energy X-ray absorptiometry (DEXA) (Day 0). Rats were then treated according to the following schedule: [0306]
  • Group 1: Vehicle controls (PBS, injected SC, [0307] Days 1, 3, and 5)
  • Group 2: OPG-Fc, single SC injection (1 mg/kg) on [0308] Day 1
  • Group 3: PTH-(1-34), SC injections on [0309] Days 1, 3, and 5, at 20 nMoles PTH/kg/injection. This represents an optimal anabolic PTH regimen.
  • Group 4: Same as [0310] group 3, but with a single OPG-Fc injection on Day 1.
  • Group 5: Single SC injection of PTH-(1-34)-Fc at 60 nMoles/kg, on [0311] Day 1. This represents a molar dose which is equivalent to the total dose of PTH-(1-34) received by group 3, but in a single injection.
  • Group 6: Same as [0312] group 5, but with a single OPG-Fc injection (SC, 1 mg/kg) on Day 1.
  • DEXA of the lumbar spine was performed again on Day 7 to evaluate changes in BMD. BMD in L3 increased modestly with a single injection of OPG-Fc, or with 3 injections of PTH-(1-34), compared to PBS-treated rats (FIG. 5). PTH-(1-34)+OPG caused a greater increase in BMD than either OPG or PTH-(1-34) alone. As a monotherapy, a single injection of PTH-(1-34)-Fc failed to increase BMD. However, a single injection of PTH-(1-34)-Fc plus a single injection of OPG-Fc caused a significant increase in BMD (FIG. 5). This result provides proof of principle that a PTH construct with extended circulating half life can be combined with a potent antiresorptive, like OPG-Fc, to create an anabolic skeletal response. The anabolic effect of a single treatment with PTH-(1-34)-Fc plus OPG-Fc was greater than that induced by multiple injections of PTH-(1-34), with or without OPG-Fc co-treatment. In conclusion, maximal gains in BMD can be achieved with infrequent injections of PTH-(1-34)-Fc+OPG-Fc, which is a superior treatment regimen compared to PTH-(1-34), which must be injected daily or every second day. [0313]
  • FIG. 5 shows the effect of PTH-Fc +OPG-Fc on bone mineral density (BMD) in the third lumbar vertebra (L3). Normal 6 month old male rats were treated with PTH-Fc or PTH or vehicle by a single SC injection. Some rats also received a single SC injection of OPG. BMD was determined 7 days later by DEXA. Data represent means±SD, n=6 rats/group. [0314]
  • EXAMPLE 2 Bioactivity of an Fc-conjugated PTH/PTHrP Receptor (PTH-R1) Antagonist ([Asn10,Leu11]PTHrP-(7-34)-Fc)
  • Introduction [0315]
  • Several disease states are associated with increased circulating levels of PTH or PTHrP. Primary and secondary hyperparathyroidism (PHPT and SHPT, respectively), are associated with increased PTH levels, while humoral hypercalcemia of malignancy (HHM) results in elevated PTHrP levels. Both proteins signal through the common PTH/PTHrP receptor (PTH-R1) to cause increases in bone resorption, renal calcium reabsorption, and renal biosynthesis of vitamin D. While bone resorption inhibitors have variable success in inhibiting osteoclastic bone resorption in these disease states, no therapy currently mitigates the intestinal and renal influence of PTH or PTHrP excess on calcemia. Agents which directly antagonize PTH or PTHrP signaling are therefore likely to have greater efficacy compared to resorption inhibitors. [0316]
  • The most studied antagonists of PTH-R1 signaling are based on amino terminal truncations. PTH-(7-34) peptides are fairly effective PTH-R1 antagonists with very mild agonist activity. Compared to PTH-(7-34), PTHrP-(7-34) peptide has greater affinity for PTH-R1 and as such is a more potent antagonist. However, PTHrP-(7-34) also has greater (but still mild) agonist activity compared to PTH-(7-34) (McKee (1990), [0317] Endocrinol. 127: 76). The optimal antagonist may combine the weaker agonism of PTH-(7-34) with the stronger antagonism of PTHrP-(7-34). Nutt et al (1990), Endocrinol. 127: 491, demonstrates that substituting Asn10 and Leu11 of PTH into the PTHrP sequence (replacing Asp10 and Lys11) results in a peptide ([Asn10,Leu11]PTHrP-(7-34)-Fc) with virtually no agonist activity but with very potent antagonist activity.
  • Like native PTH, all peptide-based PTH-R1 antagonists share the property of very short circulating half-lives (<1 h). Furthermore, the amino-terminal truncations which are required to remove receptor agonism, also significantly reduce the affinity of these peptides for PTH-R1. These properties limit the clinical potential of conventional peptide antagonists. Fc-conjugation of amino-terminally truncated PTH- or PTHrP peptides should significantly increase their circulating half life, such that continuous antagonism of PTH-R1 might be achieved with sufficient exposure to these Fc-antagonists. [0318]
  • Materials, Methods and Results [0319]
  • We have cloned, expressed and purified [Asn10,Leu11]PTHrP-(7-34)-Fc. We tested the ability of this compound to antagonize both acute and chronic hypercalcemia responses in mice. PTHrP-(1-34) was used as a calcemic agent to evaluate the acute effects of [Asn10,Leu11]PTHrP-(7-34)-Fc. Because PTHrP is the principal mediator of HHM, this study also represents a model for hypercalcemia-inducing tumors. Briefly, blood ionized calcium (BIC) was measured at baseline, and mice were then challenged with vehicle (PBS) or with PTHrP-(1-34) (0.5 mg/kg) by SC injection. Mice were then treated once SC with varying doses of [Asn10,Leu11]PTHrP-(7-34)-Fc, or with vehicle (PBS). In vehicle-treated mice challenged with PTHrP-(1-34), a transient hypercalcemic response was observed. The peak calcemic response occurred at 3 h post challenge, and persisted until at least 6 h post challenge. [Asn10,Leu11]PTHrP-(7-34)-Fc at 10 mg/kg caused a more rapid normalization of PTHrP-induced hypercalcemia compared to vehicle treatment. A dose of 30 mg/kg completely blocked the calcemic response to PTHrP-(1-34) (FIG. 6). [0320]
  • In order to test the ability of [Asn10,Leu11]PTHrP-(7-34)-Fc to antagonize more chronic hypercalcemia, we used PTH-(1-34)-Fc as a long-acting calcemic agent. This study also represents a model for primary and secondary hyperparathyroidism, diseases which are characterized by persistent elevation of PTH levels. In vehicle-treated mice, a single SC injection of PTH-(1-34)-Fc (30 mg/kg) caused a robust hypercalcemic response in normal mice, reaching a level of 2.75 mg/dl at 24 h post challenge (vs. normal control value of 1.35). A single SC injection of [Asn10,Leu11]PTHrP-(7-34)-Fc at 10-100 mg/kg caused a significant decrease in the peak hypercalcemic response to PTH-(1-34)-Fc at 24 h (FIG. 6). [0321]
  • In conclusion, we have demonstrated antagonistic activity of [Asn10,Leu11]PTHrP-(7-34)-Fc, in both acute and chronic animal models of hypercalcemia. These models employed calcemic agents based on both PTH and on PTHrP sequences. These data suggest that [Asn10,Leu11]PTHrP-(7-34)-Fc, as well as other Fc-conjugated PTH-R1 antagonists, may be effective treatment options for hyperparathyroidism, HHM, and other diseases associated with aberrant PTH-R1 signaling. [0322]
  • EXAMPLE 3 Osteogenic Properties of Fc-Conjugated and Native C-Terminally Truncated PTH Fragments
  • A. cAMP Assays [0323]
  • We tested the relative ability of PTH-Fc constructs to stimulate cAMP accumulation in rat osteoblast-like ROS 17/2.8 cells. Cultures were treated with the phosphodiesterase inhibitor IBMX to promote the accumulation of cAMP. Cultures were then challenged for 15 minutes with either vehicle (PBS), or various PTH constructs. Dose-dependent cAMP accumulation was demonstrated for all fragments. Non-Fc-conjugated PTH-(1-34) was slightly more potent than PTH-(1-31)-Fc and PTH-(1-30)-Fc (FIG. 7). These data demonstrate that Fc-conjugated PTH fragments maintain the ability to activate the AC pathway in osteoblasts. [0324]
  • B. Mouse Bioassay [0325]
  • We then tested the effects of PTH-(1-34), PTH-(1-34)-Fc, PTH-(1-31)-Fc and PTH-(1-30)-Fc in mice. Four week old male mice were injected on [0326] days 0, 5, and 10 with vehicle or with PTH fragments, by SC injection. Peripheral blood was obtained for clinical chemistry at 24, 48, and 72 h. Mice were killed on day 15, vertebrae, tibiae and femurs were harvested for histology and one tibia was collected for bone density measurements (peripheral quantitative computed tomography, pQCT). Clinical chemistry endpoints included total serum calcium, serum alkaline phosphatase (AP, a marker of osteoblast activity), and serum tartrate-resistant acid phosphatase (TRAP, a marker of osteoclast activity). For each animal, the ratio of AP:TRAP was calculated as an index of relative osteoblast activity compared to osteoclast activity. A higher AP:TRAP ratio would indicate a potentially more anabolic agent. The relatively high doses (15-fold greater than optimal anabolic doses) were selected base on previous studies which demonstrated significant changes in clinical chemistry endpoints. It was anticipated that lower doses might be required to demonstrate anabolic effects on bone density, and that antiresorptive co-treatment might also be required to achieve anabolic responses.
  • The clinical chemistry results are demonstrated in FIG. 8. Serum calcium was not significantly different at 24, 48, or 72 h after injection of 300 nmoles/kg (1.2 mg/kg) of PTH-(1-34). This result is consistent with the short half-life of the non-Fc conjugated peptide, which normally causes a transient (12 h) increase in serum calcium. In contrast, an equimolar dose of PTH-(1-34)-Fc caused a dramatic and sustained increase in serum calcium, which peaked at 24 h. PTH-(1-31)-Fc was a more potent calcemic agent, while PTH-(1-30)-Fc was the least calcemic of the 3 Fc peptides (FIG. 8A). Serum AP (osteoblast marker) was unchanged by PTH-(1-34) administration, but was significantly elevated by 300 nmoles/kg of PTH-(1-34)-Fc and by PTH-(1-31)-Fc at 72 h. PTH-(1-30)-Fc demonstrated the greatest elevation of AP, which peaked 72 h after injection of 1,000 nmoles/kg (FIG. 8B). Serum TRAP (osteoclast marker) was not significantly changed by PTH-(1-34), PTH-(1-34)-Fc, or PTH-(1-30)-Fc, but was dramatically increased by PTH-(1-31)-Fc (FIG. 8C). The calculated AP:TRAP ratios were unchanged by PTH-(1-34), and were increased over time by PTH-(1-34)-Fc. The low dose of PTH-(1-31)-Fc (100 nmoles/kg) increased AP:TRAP, while the high dose (1,000 nmoles/kg) decreased AP:TRAP. The greatest increase in AP:TRAP was realized with PTH-(1-30)-Fc (1,000 nmoles/kg) (FIG. 8D). [0327]
  • The effects of the various PTH constructs on bone mineral density (proximal tibial metaphysis) are demonstrated in FIG. 9. At the end of the 15-day study, PTH-(1-34) (300 nmoles/kg) was observed to have a modest (non-significant) anabolic effect when injected on [0328] day 0, day 5 and day 10. PTH-(1-34)-Fc (300 nmoles/kg) had no effect on bone density, nor did PTH-(1-31)-Fc at 100 nmoles/kg. Higher doses of PTH-(1-31)-Fc (300-1,000 nmoles/kg) caused significant hypercalcemia-related toxicity, and bones were not harvested from these animals for pQCT. PTH-(1-30)-Fc caused the greatest increase in bone density. There was an apparent reverse dose-response, where PTH-(1-30)-Fc at 100 nmoles/kg had the greatest effect and at 1,000 nmoles/kg had the least effect, although at all doses BMD was greater than in controls (FIG. 9). The reverse dose-response was consistent with the notion that doses employed (chosen for clinical chemistry endpoints) were 5-50 fold higher than the optimal anabolic doses. Low doses of PTH (or PTH-Fc) which fail to significantly increase serum calcium are optimal for anabolic effects. See Hock, J. M. (1992), J. Bone Min. Res. 7:65-72. In the current study, the treatment regimen with the greatest anabolic effect (PTH-(1-30)-Fc at 100 nmoles/kg) was also the only PTH-Fc treatment which failed to significantly increase serum calcium (FIG. 8A).
  • These data demonstrate the potential anabolic effects of C-terminally truncated PTH-Fc peptides. The longer half-life conferred by Fc conjugation, combined with the selective stimulation of AC/cAMP by C-terminal truncations, may explain the anabolic effect in the absence of a potent bone resorption inhibitor. It is expected that stepwise C-terminal truncation of PTH-(1-30)-Fc will reveal shorter fragments which maintain or exceed the anabolic profile of PTH-(1-30)-Fc. These fragments may be more selective at stimulating osteoblasts, and may be less calcemic, thus providing a wider therapeutic window for anabolic therapy. [0329]
  • EXAMPLE 4 PTH-Fc Treatment as a Monotherapy
  • The efficacy of PTH-(1-34)-Fc as a monotherapy was demonstrated in adult mice. Briefly, male BDF1 mice (4 months of age) were treated twice per week by subcutaneous injection with various doses of PTH-(1-34)-Fc or with vehicle (PBS). Other mice were treated daily with SC injections of PTH-(1-34) at a dose of 80 μg/kg/day (20 nmol/kg/day), a treatment regimen which is optimal for increasing bone mass in rodents (M. Gunness-Hey and J. M. Hock, [0330] Metab. Bone Dis. & Rel. Res. 5:177-181, 1984). After 3 weeks, mice were sacrificed and tibiae were analyzed for bone mineral density (BMD) via pQCT (FIG. 10).
  • Total tibial BMD and cancelled BMD were both significantly increased by daily PTH-(1-34) injections compared to vehicle-treated controls (FIG. 1, two-way ANOVA, p<0.05). Twice-weekly injections of PTH-(1-34)-Fc caused dose-dependent increases in both total and cancellous BMD which, at the two highest doses (50 and 150 nmol/kg), were significantly greater than the effects of either vehicle or daily PTH-(1-34). Cortical BMD in the tibia was not significantly enhanced by daily PTH-(1-34) treatments. Twice-weekly PTH-(1-34)-Fc caused a dose-dependent increase in cortical BMD which at the highest dose was signficantly greater than that observed in mice treated with vehicle or with daily PTH-(1-34) (p<0.05). [0331]
  • Twice-weekly PTH-(1-34)-Fc also effectively increased BMD as a monotherapy in aged ovariectomized (OVX) rats. Sprague Dawley rats were OVX'd at 3 months of age and allowed to lose bone for 11 months. Other rats were sham-operated and treated twice per week with vehicle (PBS). OVX rats were treated twice per week with SC injections of either vehicle or the bisphosphonate APD (pamidronate, 0.5 mg/kg), or with PTH-(1-34)-Fc (50 nmol/kg) or with APD+PTH-(1-34)-Fc. BMD was determined weekly via dual energy X-ray absorptiometry (DEXA). Rats were sacrificed after 4 weeks of treatment. At the start of treatment, OVX rats had significant reductions in BMD at all skeletal sites analyzed, compared to vehicle-treated OVX rats (FIG. 11, p<0.05, 2-way ANOVA). APD alone did not significantly increase BMD at any skeletal site compared to vehicle-treated OVX rats. PTH-(1-34)-Fc alone caused a significant increase in BMD at the femoral metaphysis after 4 weeks of treatment (p<0.05). Treatment of OVX rats with PTH+ABD was associated with an earlier significant increase in BMD at this site (3 weeks). The combination of APD+PTH-(1-34)-Fc also caused significant BMD increases at the lumbar vertebrae and at the femoral metaphysis (p<0.05). PTH-(1-34)-Fc alone caused a mild and transient hypercalcemic response which resolved spontaneously after [0332] day 10 despite continued treatments. The co-administration of APD completely blocked the calcemic effect of PTH-(1-34)-Fc.
  • These data suggest that PTH-(1-34)-Fc is an effective anabolic agent when used as a monotherapy in both adult mice and aged OVX rats. We have also demonstrated that the addition of an antiresorptive agent (APD) to PTH-(1-34)-Fc was associated with similar or more rapid increases in BMD in aged OVX rats. Co-administration of APD also blocked the transient hypercalcemic response produced by PTH-(1-34)-Fc, which suggests that the therapeutic index of PTH-(1-34)-Fc could be significantly improved by co-administering an effective antiresorptive agent. [0333]
  • The invention now being fully described, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto, without departing from the spirit and scope of the invention as set forth herein. [0334]
    Abbreviations
    AC adenylate cyclase
    AP alkaline phosphatase
    BMD bone mineral density
    cAMP cyclic adenosine monophosphate
    DEXA dual-energy X-ray absorptiometry
    HHM humoral hypercalcemia of malignancy
    OPG osteoprotegerin
    OVX ovariectomized
    PBS phosphate-buffered saline
    pQCT peripheral quantitative computed tomography
    PTH parathyroid hormone
    PTHrP parathyroid hormone-related protein
    TRAP tartrate-resistant acid phosphatase
  • [0335]
  • 1 170 1 684 DNA Homo sapiens CDS (1)..(684) 1 atg gac aaa act cac aca tgt cca cct tgt cca gct ccg gaa ctc ctg 48 Met Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu 1 5 10 15 ggg gga ccg tca gtc ttc ctc ttc ccc cca aaa ccc aag gac acc ctc 96 Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu 20 25 30 atg atc tcc cgg acc cct gag gtc aca tgc gtg gtg gtg gac gtg agc 144 Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser 35 40 45 cac gaa gac cct gag gtc aag ttc aac tgg tac gtg gac ggc gtg gag 192 His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 50 55 60 gtg cat aat gcc aag aca aag ccg cgg gag gag cag tac aac agc acg 240 Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr 65 70 75 80 tac cgt gtg gtc agc gtc ctc acc gtc ctg cac cag gac tgg ctg aat 288 Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn 85 90 95 ggc aag gag tac aag tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc 336 Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro 100 105 110 atc gag aaa acc atc tcc aaa gcc aaa ggg cag ccc cga gaa cca cag 384 Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln 115 120 125 gtg tac acc ctg ccc cca tcc cgg gat gag ctg acc aag aac cag gtc 432 Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val 130 135 140 agc ctg acc tgc ctg gtc aaa ggc ttc tat ccc agc gac atc gcc gtg 480 Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val 145 150 155 160 gag tgg gag agc aat ggg cag ccg gag aac aac tac aag acc acg cct 528 Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro 165 170 175 ccc gtg ctg gac tcc gac ggc tcc ttc ttc ctc tac agc aag ctc acc 576 Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr 180 185 190 gtg gac aag agc agg tgg cag cag ggg aac gtc ttc tca tgc tcc gtg 624 Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val 195 200 205 atg cat gag gct ctg cac aac cac tac acg cag aag agc ctc tcc ctg 672 Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu 210 215 220 tct ccg ggt aaa 684 Ser Pro Gly Lys 225 2 228 PRT Homo sapiens 2 Met Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu 1 5 10 15 Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu 20 25 30 Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser 35 40 45 His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 50 55 60 Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr 65 70 75 80 Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn 85 90 95 Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro 100 105 110 Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln 115 120 125 Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val 130 135 140 Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val 145 150 155 160 Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro 165 170 175 Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr 180 185 190 Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val 195 200 205 Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu 210 215 220 Ser Pro Gly Lys 225 3 21 PRT Artificial Sequence PTH/PTHrP 3 Xaa His Xaa Xaa Xaa Lys Xaa Xaa Xaa Xaa Xaa Xaa Arg Xaa Xaa Xaa 1 5 10 15 Xaa Xaa Xaa Xaa Xaa 20 4 22 PRT Artificial Sequence PTH/PTHrP 4 Xaa Xaa His Asn Leu Xaa Lys His Leu Xaa Ser Xaa Xaa Arg Xaa Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu 20 5 21 PRT Artificial Sequence PTH/PTHrP 5 Leu His Xaa Xaa Xaa Lys Ser Ile Xaa Xaa Leu Arg Arg Arg Phe Xaa 1 5 10 15 Leu His His Leu Ile 20 6 8 PRT Artificial Sequence Preferred linker 6 Gly Gly Gly Lys Gly Gly Gly Gly 1 5 7 8 PRT Artificial Sequence Preferred linker 7 Gly Gly Gly Asn Gly Ser Gly Gly 1 5 8 8 PRT Artificial Sequence Preferred linker 8 Gly Gly Gly Cys Gly Gly Gly Gly 1 5 9 5 PRT Artificial Sequence Preferred linker 9 Gly Pro Asn Gly Gly 1 5 10 84 PRT Homo sapiens 10 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe Val Ala Leu Gly Ala Pro Leu Ala Pro Arg Asp Ala Gly Ser 35 40 45 Gln Arg Pro Arg Lys Lys Glu Asp Asn Val Leu Val Glu Ser His Glu 50 55 60 Lys Ser Leu Gly Glu Ala Asp Lys Ala Asp Val Asn Val Leu Thr Lys 65 70 75 80 Ala Lys Ser Gln 11 84 PRT Rattus rattus 11 Ala Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Ala 1 5 10 15 Ser Val Glu Arg Met Gln Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe Val Ser Leu Gly Val Gln Met Ala Ala Arg Glu Gly Ser Tyr 35 40 45 Gln Arg Pro Thr Lys Lys Glu Asp Asn Val Leu Val Asp Gly Asn Ser 50 55 60 Lys Ser Leu Gly Glu Gly Asp Lys Ala Asp Val Asp Val Leu Val Lys 65 70 75 80 Ala Lys Ser Gln 12 78 PRT Homo sapiens 12 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Phe Val Ala Leu Gly 20 25 30 Ala Pro Leu Ala Pro Arg Asp Ala Gly Ser Gln Arg Pro Arg Lys Lys 35 40 45 Glu Asp Asn Val Leu Val Glu Ser His Glu Lys Ser Leu Gly Glu Ala 50 55 60 Asp Lys Ala Asp Val Asn Val Leu Thr Lys Ala Lys Ser Gln 65 70 75 13 44 PRT Homo sapiens 13 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe Val Ala Leu Gly Ala Pro Leu Ala Pro Arg 35 40 14 38 PRT Homo sapiens 14 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe Val Ala Leu Gly 35 15 37 PRT Homo sapiens 15 Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn Ser 1 5 10 15 Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn 20 25 30 Phe Val Ala Leu Gly 35 16 34 PRT Homo sapiens 16 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 17 34 PRT Artificial Sequence modified human PTH 17 Ser Val Ser Glu Ile Gln Leu Met His Asn Arg Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 18 34 PRT Artificial Sequence modified human PTH 18 Ser Val Ser Glu Ile Gln Leu Met His Asn Lys Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 19 34 PRT Artificial Sequence modified human PTH 19 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Arg Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 20 34 PRT Artificial Sequence modified human PTH 20 Tyr Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 21 34 PRT Artificial Sequence modified human PTH 21 Ser Val Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Tyr 22 34 PRT Artificial Sequence bovine 22 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 23 34 PRT Artificial Sequence modified bovine PTH 23 Ala Val Ser Glu Ile Gln Phe Leu His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Tyr 24 34 PRT Artificial Sequence porcine 24 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 25 34 PRT Rattus rattus 25 Ala Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Ala 1 5 10 15 Ser Val Glu Arg Met Gln Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 26 34 PRT Artificial Sequence modified rat PTH 26 Ala Val Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Ala 1 5 10 15 Ser Val Glu Arg Leu Gln Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Tyr 27 31 PRT Homo sapiens 27 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val 20 25 30 28 31 PRT Artificial Sequence modified human PTH 28 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Leu Leu Gln Asp Val 20 25 30 29 32 PRT Artificial Sequence PTH 29 Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Asn Ser Leu 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 30 30 32 PRT Artificial Sequence bovine 30 Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Phe 20 25 30 31 32 PRT Artificial Sequence modified bovine PTH 31 Ser Glu Ile Gln Phe Leu His Asn Leu Gly Lys His Leu Ser Ser Leu 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 30 32 28 PRT Homo sapiens 32 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Phe 20 25 33 28 PRT Artificial Sequence modified human PTH 33 Leu Leu His Asn Leu Gly Lys His Leu Asn Ser Leu Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 34 28 PRT Artificial Sequence bovine PTH 34 Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Phe 20 25 35 28 PRT Artificial Sequence modified bovine PTH 35 Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 36 28 PRT Artificial Sequence modified bovine PTH 36 Phe Leu His Asn Leu Gly Lys His Leu Ser Ser Leu Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 37 5 PRT Artificial Sequence modified bovine PTH 37 Cys Asn Gly Arg Cys 1 5 38 27 PRT Artificial Sequence modified bovine PTH 38 Phe Met His Asn Leu Lys His Leu Ser Ser Met Glu Arg Val Glu Trp 1 5 10 15 Leu Arg Lys Lys Leu Gln Asp Val His Asn Tyr 20 25 39 30 PRT Homo sapiens 39 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 40 30 PRT Artificial Sequence modified human PTH 40 Ser Val Ser Glu Ile Gln Leu Met His Asn Arg Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 41 30 PRT Artificial Sequence modified human PTH 41 Ser Val Ser Glu Ile Gln Leu Met His Asn Lys Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 42 30 PRT Artificial Sequence modified human PTH 42 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Arg Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 43 30 PRT Artificial Sequence modified human PTH 43 Tyr Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 44 30 PRT Artificial Sequence modified human PTH 44 Ser Val Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 45 30 PRT Artificial Sequence bovine 45 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 46 30 PRT Artificial Sequence modified bovine PTH 46 Ala Val Ser Glu Ile Gln Phe Leu His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 47 30 PRT Artificial Sequence porcine PTH 47 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Leu Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 48 30 PRT Artificial Sequence rat PTH 48 Ala Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Ala 1 5 10 15 Ser Val Glu Arg Met Gln Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 49 30 PRT Artificial Sequence modified rat PTH 49 Ala Val Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Ala 1 5 10 15 Ser Val Glu Arg Leu Gln Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 50 30 PRT Artificial Sequence modified human PTH 50 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Leu Leu Gln Asp 20 25 30 51 29 PRT Homo sapiens 51 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln 20 25 52 28 PRT Homo sapiens 52 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu 20 25 53 28 PRT Artificial Sequence modified PTH 53 Ser Glu Ile Gln Leu Leu His Asn Leu Gly Lys His Leu Asn Ser Leu 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 54 28 PRT Artificial Sequence bovine 54 Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 55 28 PRT Artificial Sequence modified bovine PTH 55 Ser Glu Ile Gln Phe Leu His Asn Leu Gly Lys His Leu Ser Ser Leu 1 5 10 15 Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 56 24 PRT Homo sapiens 56 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 57 24 PRT Artificial Sequence modified human PTH 57 Leu Leu His Asn Leu Gly Lys His Leu Asn Ser Leu Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 58 24 PRT Artificial Sequence bovine 58 Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 59 24 PRT Artificial Sequence modified bovine PTH 59 Phe Leu His Asn Leu Gly Lys His Leu Ser Ser Leu Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 60 24 PRT Artificial Sequence modified bovine PTH 60 Phe Leu His Asn Leu Trp Lys His Leu Ser Ser Leu Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 61 24 PRT Artificial Sequence modified bovine PTH 61 Phe Met His Asn Leu Lys Trp His Leu Ser Ser Met Glu Arg Val Glu 1 5 10 15 Trp Leu Arg Lys Lys Leu Gln Asp 20 62 86 PRT Homo sapiens 62 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu Ile His 20 25 30 Thr Ala Glu Ile Arg Ala Thr Ser Glu Val Ser Pro Asn Ser Lys Pro 35 40 45 Ser Pro Asn Thr Lys Asn His Pro Val Arg Phe Gly Ser Asp Asp Glu 50 55 60 Gly Arg Tyr Leu Thr Gln Glu Thr Asn Lys Val Glu Thr Tyr Lys Glu 65 70 75 80 Gln Pro Leu Lys Thr Pro 85 63 34 PRT Homo sapiens 63 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu Ile His 20 25 30 Thr Ala 64 36 PRT Artificial Sequence modified human PTHrP 64 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu Ile His 20 25 30 Thr Ala Glu Tyr 35 65 36 PRT Artificial Sequence modified human PTHrP 65 Ala Val Ser Glu Ile Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Trp Leu His His Leu Ile Ala Glu Ile His 20 25 30 Thr Ala Glu Tyr 35 66 35 PRT Artificial Sequence modified human PTHrP 66 Tyr Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile 1 5 10 15 Gln Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu Ile 20 25 30 His Thr Ala 35 67 33 PRT Artificial Sequence modified human PTHrP 67 Ala Val Ser Glu His Gln Leu Leu His Asn Leu Lys Ser Ile Gln Asp 1 5 10 15 Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu Ile His Thr 20 25 30 Ala 68 28 PRT Homo sapiens 68 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 69 28 PRT Artificial Sequence modified human PTHrP 69 Leu Leu His Asn Leu Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 70 28 PRT Artificial Sequence modified PTHrP 70 Leu Leu His Asp Lys Gly Lys Ser Ile Asn Leu Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 71 28 PRT Artificial Sequence modified human PTHrP 71 Leu Leu His Asp Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 72 28 PRT Artificial Sequence modified PTHrP 72 Leu Leu His Asn Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 73 27 PRT Artificial Sequence modified PTHrP 73 Leu His Asn Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 74 27 PRT Artificial Sequence modified PTHrP 74 Leu His Asn Leu Phe Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 75 28 PRT Artificial Sequence modified human PTHrP 75 Leu Leu His Asn Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 76 30 PRT Homo sapiens 76 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu 20 25 30 77 30 PRT Artificial Sequence modified human PTHrP 77 Ala Val Ser Glu Ile Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Trp Leu His His Leu Ile Ala Glu 20 25 30 78 31 PRT Artificial Sequence human PTHrP with non-human N-terminal peptide 78 Tyr Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile 1 5 10 15 Gln Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu 20 25 30 79 30 PRT Artificial Sequence modified human PTHrP 79 Ala Val Ser Glu His Gln Leu Leu His Asn Leu Phe Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Phe Phe Leu His His Leu Ile Ala Glu 20 25 30 80 24 PRT Homo sapiens 80 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu 20 81 24 PRT Artificial Sequence modified human PTHrP 81 Leu Leu His Asn Leu Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu 20 82 24 PRT Artificial Sequence modified PTHrP 82 Leu Leu His Asp Lys Gly Lys Ser Ile Asn Leu Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu 20 83 23 PRT Artificial Sequence modified human PTHrP 83 Leu Leu His Asp Leu Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu 20 84 23 PRT Artificial Sequence modified PTHrP 84 Leu Leu His Asn Leu Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu 20 85 23 PRT Artificial Sequence modified PTHrP 85 Leu His Asn Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu 20 86 23 PRT Artificial Sequence modified PTHrP 86 Leu His Asn Leu Phe Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe Phe 1 5 10 15 Leu His His Leu Ile Ala Glu 20 87 24 PRT Artificial Sequence modified human PTHrP 87 Leu Leu His Asn Leu Trp Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu 20 88 34 PRT Artificial Sequence modified human PTH 88 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Asn Phe 89 34 PRT Artificial Sequence modified human PTH 89 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Lys Lys Leu His 20 25 30 Asn Phe 90 34 PRT Artificial Sequence modified human PTH 90 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu His 20 25 30 Asn Phe 91 34 PRT Artificial Sequence modified human PTH 91 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ser Leu Leu Ser Ser Leu Leu Ser Ser Leu His 20 25 30 Asn Phe 92 34 PRT Artificial Sequence modified human PTH 92 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ala Phe Tyr Asp Lys Val Ala Glu Lys Leu His 20 25 30 Asn Phe 93 28 PRT Artificial Sequence modified human PTH 93 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Glu Lys Leu His Asn Phe 20 25 94 28 PRT Artificial Sequence modified human PTH 94 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Lys Lys Leu His Asn Phe 20 25 95 28 PRT Artificial Sequence modified human PTH 95 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ala 1 5 10 15 Leu Ala Glu Ala Leu Ala Glu Ala Leu His Asn Phe 20 25 96 28 PRT Artificial Sequence modified human PTH 96 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ser 1 5 10 15 Leu Leu Ser Ser Leu Leu Ser Ser Leu His Asn Phe 20 25 97 28 PRT Artificial Sequence modified human PTH 97 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ala 1 5 10 15 Phe Tyr Asp Lys Val Ala Glu Lys Leu His Asn Phe 20 25 98 34 PRT Artificial Sequence modified human PTHrP 98 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr Ala 99 34 PRT Artificial Sequence modified human PTHrP 99 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Lys Lys Leu His 20 25 30 Thr Ala 100 34 PRT Artificial Sequence modified human PTHrP 100 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu His 20 25 30 Thr Ala 101 34 PRT Artificial Sequence modified human PTHrP 101 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ser Leu Leu Ser Ser Leu Leu Ser Ser Leu His 20 25 30 Thr Ala 102 34 PRT Artificial Sequence modified human PTHrP 102 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Phe Tyr Asp Lys Val Ala Glu Lys Leu His 20 25 30 Thr Ala 103 28 PRT Artificial Sequence modified human PTHrP 103 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Glu Lys Leu His Thr Ala 20 25 104 28 PRT Artificial Sequence modified human PTHrP 104 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Lys Lys Leu His Thr Ala 20 25 105 28 PRT Artificial Sequence modified human PTHrP 105 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ala 1 5 10 15 Leu Ala Glu Ala Leu Ala Glu Ala Leu His Thr Ala 20 25 106 28 PRT Artificial Sequence modified human PTHrP 106 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ser 1 5 10 15 Leu Leu Ser Ser Leu Leu Ser Ser Leu His Thr Ala 20 25 107 28 PRT Artificial Sequence modified human PTHrP 107 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ala 1 5 10 15 Phe Tyr Asp Lys Val Ala Glu Lys Leu His Thr Ala 20 25 108 34 PRT Artificial Sequence modified human PTHrP 108 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Arg Lys Leu His 20 25 30 Thr Ala 109 34 PRT Artificial Sequence modified human PTHrP 109 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr Ser 110 37 PRT Artificial Sequence modified human PTHrP 110 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr Ala Gly Arg Arg 35 111 34 PRT Artificial Sequence modified human PTHrP 111 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu Lys 20 25 30 Glu Leu 112 34 PRT Artificial Sequence modified human PTHrP 112 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Ala Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr Ala 113 34 PRT Artificial Sequence modified human PTHrP 113 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Ala Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr Ala 114 34 PRT Artificial Sequence modified human PTHrP 114 Ala Val Ser Glu Ala Gln Leu Leu His Asp Leu Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Ala Leu 115 34 PRT Artificial Sequence modified human PTHrP 115 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Glu Arg Leu His 20 25 30 Thr Ala 116 33 PRT Artificial Sequence modified human PTHrP 116 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Arg Ser Ile Gln 1 5 10 15 Asp Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Glu Arg Leu His Thr 20 25 30 Ala 117 34 PRT Artificial Sequence modified human PTHrP 117 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Lys Arg Leu His 20 25 30 Thr Ala 118 34 PRT Artificial Sequence modified human PTHrP 118 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Arg Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Lys Arg Leu His 20 25 30 Thr Ala 119 34 PRT Artificial Sequence modified human PTHrP 119 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu His 20 25 30 Thr Ala 120 34 PRT Artificial Sequence modified human PTHrP 120 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ser Leu Leu Ser Ser Leu Leu Ser Ser Leu His 20 25 30 Thr Ala 121 34 PRT Artificial Sequence modified human PTHrP 121 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Phe Tyr Asp Lys Val Ala Glu Lys Leu His 20 25 30 Thr Ala 122 34 PRT Artificial Sequence modified human PTHrP 122 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Asn Tyr 123 34 PRT Artificial Sequence modified human PTHrP 123 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Asn Tyr 124 30 PRT Artificial Sequence modified human PTH 124 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 125 30 PRT Artificial Sequence modified human PTH 125 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Lys Lys 20 25 30 126 30 PRT Artificial Sequence modified human PTH 126 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ala Leu Ala Glu Ala Leu Ala Glu Ala 20 25 30 127 30 PRT Artificial Sequence modified human PTH 127 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ser Leu Leu Ser Ser Leu Leu Ser Ser 20 25 30 128 34 PRT Artificial Sequence modified human PTH 128 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Ala Phe Tyr Asp Lys Val Ala Glu Lys Leu His 20 25 30 Asn Phe 129 24 PRT Artificial Sequence modified human PTH 129 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Glu Lys 20 130 24 PRT Artificial Sequence modified human PTH 130 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Lys Lys 20 131 24 PRT Artificial Sequence modified human PTH 131 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ala 1 5 10 15 Leu Ala Glu Ala Leu Ala Glu Ala 20 132 24 PRT Artificial Sequence modified human PTH 132 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ser 1 5 10 15 Leu Leu Ser Ser Leu Leu Ser Ser 20 133 24 PRT Artificial Sequence modified human PTH 133 Leu Met His Asn Leu Gly Lys His Leu Asn Ser Met Glu Arg Val Ala 1 5 10 15 Phe Tyr Asp Lys Val Ala Glu Lys 20 134 30 PRT Artificial Sequence modified human PTHrP 134 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 135 30 PRT Artificial Sequence modified human PTHrP 135 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Lys Lys 20 25 30 136 30 PRT Artificial Sequence modified human PTHrP 136 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Leu Ala Glu Ala Leu Ala Glu Ala 20 25 30 137 30 PRT Artificial Sequence modified human PTHrP 137 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ser Leu Leu Ser Ser Leu Leu Ser Ser 20 25 30 138 30 PRT Artificial Sequence modified human PTHrP 138 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Phe Tyr Asp Lys Val Ala Glu Lys 20 25 30 139 24 PRT Artificial Sequence modified human PTHrP 139 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Glu Lys 20 140 24 PRT Artificial Sequence modified human PTHrP 140 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Glu 1 5 10 15 Leu Leu Glu Lys Leu Leu Lys Lys 20 141 24 PRT Artificial Sequence modified human PTHrP 141 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ala 1 5 10 15 Leu Ala Glu Ala Leu Ala Glu Ala 20 142 24 PRT Artificial Sequence modified human PTHrP 142 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ser 1 5 10 15 Leu Leu Ser Ser Leu Leu Ser Ser 20 143 24 PRT Artificial Sequence modified human PTHrP 143 Leu Leu His Asp Lys Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Ala 1 5 10 15 Phe Tyr Asp Lys Val Ala Glu Lys 20 144 30 PRT Artificial Sequence modified human PTHrP 144 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Arg Lys 20 25 30 145 30 PRT Artificial Sequence modified human PTHrP 145 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 146 33 PRT Artificial Sequence modified human PTHrP 146 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys Leu His 20 25 30 Thr 147 30 PRT Artificial Sequence modified human PTHrP 147 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 148 30 PRT Artificial Sequence modified human PTHrP 148 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Ala Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 149 30 PRT Artificial Sequence modified human PTHrP 149 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Ala Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 150 30 PRT Artificial Sequence modified human PTHrP 150 Ala Val Ser Glu Ala Gln Leu Leu His Asp Leu Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 151 30 PRT Artificial Sequence modified human PTHrP 151 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Glu Arg 20 25 30 152 29 PRT Artificial Sequence modified human PTHrP 152 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Arg Ser Ile Gln 1 5 10 15 Asp Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Glu Arg 20 25 153 30 PRT Artificial Sequence modified human PTHrP 153 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Lys Arg 20 25 30 154 30 PRT Artificial Sequence modified human PTHrP 154 Ala Val Ser Glu His Gln Leu Leu His Asp Arg Gly Arg Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Glu Leu Leu Glu Arg Leu Leu Lys Arg 20 25 30 155 30 PRT Artificial Sequence modified human PTHrP 155 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Leu Ala Glu Ala Leu Ala Glu Ala 20 25 30 156 30 PRT Artificial Sequence modified human PTHrP 156 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ser Leu Leu Ser Ser Leu Leu Ser Ser 20 25 30 157 30 PRT Artificial Sequence modified human PTHrP 157 Ala Val Ser Glu His Gln Leu Leu His Asp Lys Gly Lys Ser Ile Gln 1 5 10 15 Asp Leu Arg Arg Arg Ala Phe Tyr Asp Lys Val Ala Glu Lys 20 25 30 158 30 PRT Artificial Sequence modified human PTHrP 158 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Glu Arg Val Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 159 30 PRT Artificial Sequence modified human PTHrP 159 Ala Val Ser Glu Ile Gln Phe Met His Asn Leu Gly Lys His Leu Ser 1 5 10 15 Ser Met Arg Arg Arg Glu Leu Leu Glu Lys Leu Leu Glu Lys 20 25 30 160 39 PRT Artificial Sequence TIP39 160 Ser Leu Ala Leu Ala Asp Asp Ala Ala Phe Arg Glu Arg Ala Arg Leu 1 5 10 15 Leu Ala Ala Leu Glu Arg Arg His Trp Leu Asn Ser Tyr Met His Lys 20 25 30 Leu Leu Val Leu Asp Ala Pro 35 161 34 PRT Artificial Sequence Preferred embodiments - PTH 161 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 162 34 PRT Artificial Sequence Preferred embodiments - PTH 162 Ser Val Ser Glu Ile Gln Leu Met His Asn Arg Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 163 34 PRT Artificial Sequence Preferred embodiments - PTH 163 Ser Val Ser Glu Ile Gln Leu Met His Asn Lys Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 164 34 PRT Artificial Sequence Preferred embodiments - PTH 164 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Arg Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val His 20 25 30 Asn Phe 165 31 PRT Artificial Sequence Preferred embodiments - PTH 165 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp Val 20 25 30 166 30 PRT Artificial Sequence Preferred embodiments - PTH 166 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln Asp 20 25 30 167 29 PRT Artificial Sequence Preferred embodiments - PTH 167 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu Gln 20 25 168 28 PRT Artificial Sequence Preferred embodiments - PTH 168 Ser Val Ser Glu Ile Gln Leu Met His Asn Leu Gly Lys His Leu Asn 1 5 10 15 Ser Met Glu Arg Val Glu Trp Leu Arg Lys Lys Leu 20 25 169 28 PRT Artificial Sequence Preferred embodiments - PTHrP 169 Leu Leu His Asn Leu Gly Lys Ser Ile Gln Asp Leu Arg Arg Arg Phe 1 5 10 15 Phe Leu His His Leu Ile Ala Glu Ile His Thr Ala 20 25 170 39 PRT Artificial Sequence Preferred embodiments - TIP39 170 Ser Leu Ala Leu Ala Asp Asp Ala Ala Phe Arg Glu Arg Ala Arg Leu 1 5 10 15 Leu Ala Ala Leu Glu Arg Arg His Trp Leu Asn Ser Tyr Met His Lys 20 25 30 Leu Leu Val Leu Asp Ala Pro 35

Claims (79)

What is claimed is:
1. A composition of matter of the formula
P1-(L1)a-F1
and multimers thereof, wherein:
F1 is a vehicle and is attached at the C-terminus of P1-(L1)a;
P1 is a PTH/PTHrP modulating domain;
L1 is a linker; and
a is 0 or 1.
2. The composition of matter of claim 1 of the formulae
P1-F1.
3. The composition of matter of claim 1, wherein F1 is an Fc domain.
4. The composition of matter of claim 1 wherein F1 is an IgG Fc domain.
5. The composition of matter of claim 1 wherein F1 is an IgG1 Fc domain.
6. The composition of matter of claim 1 wherein F1 comprises the sequence of SEQ ID NO: 2.
7. The composition of matter of claim 1 wherein the PTH/PTHrP modulating domain is of the formula
XNHX10X11X12KX14X15X16X17X18X19RX21 (SEQ ID NO: 3) X22X23X24X25X26X27X28XC
wherein:
XN is absent or is X3X4X5X6X7, X2X3X4X5X6X7, X1X2X3X4X5X6X7, or YX1X2X3X4X5X6X7;
X1 through X7, X10, X11, X12, X14 through X28 are each independently amino acid residues;
XC is absent or is X29, X29X30, X29X30X31, X29X30X31X32, X29X30X31X32X33, X29X30X31X32X33X34, X29X30X31X32X33X34X35, or X29X30X31X32X33X34X35X36;
X29 through X36 are each independently amino acid residues.
8. The composition of matter of claim 7, wherein:
XN is X1X2X3X4X5X6X7;
X1 is a hydrophilic or nonfunctional residue;
X2 is V;
X3 is S;
X4 is E;
X5 is a nonfunctional or basic residue;
X6 is Q;
X7 is L;
X10 is an acidic or hydrophilic residue;
X11 is a nonfunctional or basic residue;
X12 is a nonfunctional residue;
X14 is a basic or hydrophilic residue;
X15 is a nonfunctional residue;
X16 is a nonfunctional or hydrophilic residue;
X17 is an acidic, hydrophilic, or nonfunctional residue;
X18 is a nonfunctional residue;
X19 is an acidic or basic residue;
X21 is a nonfunctional or basic residue;
X22 is a hydrophilic, acidic, or aromatic residue;
X23 is an aromatic or lipophilic residue;
X24 is a lipophilic residue (L preferred);
X25 is a hydrophilic or basic residue;
X26 is a hydrophilic or basic residue;
X27 is a lipophilic, basic, or nonfunctional residue; and
X28 is a lipophilic or nonfunctional residue.
9. The composition of matter of claim 8, wherein:
XC is X29X30X31X32X33X34;
X29 is a hydrophilic or nonfunctional residue;
X30 is a hydrophilic or acidic residue;
X31 is a lipophilic or nonfunctional residue;
X32 is H;
X33 is a hydrophilic residue; and
X34 is a nonfunctional or aromatic residue.
10. The composition of matter of claim 8, wherein:
XC is X29X30X31;
X29 is a hydrophilic or nonfunctional residue;
X30 is a hydrophilic or acidic residue; and
X31 is a lipophilic or nonfunctional residue.
11. The composition of matter of claim 8, wherein:
XC is X29X30;
X29 is a hydrophilic or nonfunctional residue; and
X30 is a hydrophilic or acidic residue.
12. The composition of matter of claim 8, wherein:
XC is X29; and;
X29 is a hydrophilic or nonfunctional residue.
13. The composition of matter of claim 8, wherein XC is absent.
14. The composition of matter of claim 8, wherein:
X1 is A, S or Y;
X5 is H or I;
X10 is N or D;
X11 is L, R, or K;
X12 is G, F, or W;
X14 is H or S;
X15 is L or I;
X16 is Q, N, S, or A;
X17 is S, D, or L;
X18 is M, L, V or Nle;
X19 is E or R;
X21 is V, M, R, or Nle;
X22 is E or F;
X23 is W or F;
X25 is R or H;
X26 is K or H;
X27 is K or L; and
X28 is L or I.
15. The composition of matter of claim 14, wherein:
XC is X29X30X31X32X33X34;
X29 is Q or A;
X30 is D or E;
X31 is V or I;
X33 is N or T; and
X34 is A, F or Y.
16. The composition of matter of claim 14, wherein:
XC is X29X30X31;
X29 is Q or A;
X30 is D or E; and
X31 is V or I;
17. The composition of matter of claim 14, wherein:
XC is X29X30;
X29 is Q or A; and
X30 is D or E.
18. The composition of matter of claim 14, wherein:
XC is X29; and;
X29 is Q or A.
19. The composition of matter of claim 14, wherein XC is absent.
20. The composition of matter of claim 1, wherein the PTH/PTHrP modulating domain is of the formula
JNJ7J8HNJ11J12KHLJ16 (SEQ ID NO: 4) SJ18J19RJ21EWLRKKLJC
wherein:
JN is absent or is selected from J1J2J3J4J5J6, J2J3J4J5J6, J3J4J5J6;
J1 is an amino acid residue;
J2 is an amino acid residue;
J3 is an amino acid residue;
J4 is an amino acid residue;
J5 is an amino acid residue;
J6 is an amino acid residue;
J7 is an amino acid residue;
J8 is an amino acid residue;
J11 is a nonfunctional or basic residue;
J12 is an amino acid residue;
J16 is an amino acid residue;
J18 is an amino acid residue;
J19 is an acidic or basic residue;
J21 is an amino acid residue;
JC is absent or is J29, J29J30, J29J30J31, J29J30J31J32, J29J30J31J32J33, J29J30J31J32J33J34; and
J29 is an amino acid residue;
J30 is an amino acid residue;
J31 is an amino acid residue;
J32 is an amino acid residue;
J33 is an amino acid residue;
J34 is an amino acid residue.
21. The composition of matter of claim 20, wherein:
JN is J1J2J3J4J5J6;
J1 is a nonfunctional or aromatic residue;
J2 is a nonfunctional residue;
J3 is a hydrophilic residue;
J4 is an acidic residue;
J5 is a nonfunctional residue;
J6 is a basic residue;
J7 is a nonfunctional or aromatic residue;
J8 is a nonfunctional residue;
J11 is a basic or a nonfunctional residue;
J12 is a nonfunctional or aromatic residue;
J16 is a nonfunctional or hydrophilic residue;
J18 is a nonfunctional residue;
J19 is an acidic or basic residue; and
J21 is a nonfunctional residue;
JC is J29J30J31J32J33J34;
J29 is a hydrophilic or nonfunctional residue;
J30 is a hydrophilic or acidic residue;
J31 is a lipophilic or nonfunctional residue;
J32 is a basic residue;
J33 is an acidic residue; and
J34 is an aromatic residue.
22. The composition of matter of claim 21, wherein:
J1 is A, S or Y;
J2 is V;
J3 is S;
J4 is E;
J5 is I;
J6 is Q;
J7 is L or F;
J8 is M or Nle;
J11 is L, R, or K;
J12 is G or W;
J16 is N, S, or A;
J18 is M, Nle, L, or V;
J19 is E or R;
J21 is V, M, or Nle;
J29 is Q or A;
J30 is D or E;
J31 is V or I;
J32 is H;
J33 is N; and
J34is F or Y.
23. The composition of matter of claim 20, wherein:
JN is J1J2J3J4J5J6;
J1 is a nonfunctional or aromatic residue;
J2 is a nonfunctional residue;
J3 is a hydrophilic residue;
J4 is an acidic residue;
J5 is a nonfunctional residue;
J6 is a basic residue;
J7 is a nonfunctional or aromatic residue;
J8 is a nonfunctional residue;
J11 is a basic or a nonfunctional residue;
J12 is a nonfunctional or aromatic residue;
J16 is a nonfunctional or hydrophilic residue;
J18 is a nonfunctional residue;
J19 is an acidic or basic residue;
J21 is a nonfunctional residue;
JC is J29J30J31;
J29 is a hydrophilic or nonfunctional residue;
J30 is a hydrophilic or acidic residue; and
J31 is a lipophilic or nonfunctional residue.
24. The composition of matter of claim 23, wherein:
J1 is A, S or Y;
J2 is V;
J3 is S;
J4is E;
J5 is I;
J6 is Q;
J7is L or F;
J8 is M or Nle;
J11 is L, R, or K;
J12 is G or W;
J16 is N, S, or A;
J18 is M, Nle, L, or V;
J19 is E or R;
J21 is V, M, or Nle;
J29 is Q or A;
J30 is D or E; and
J31 is V or I.
25. The composition of matter of claim 20, wherein:
JN is J1J2J3J4J5J6;
J1 is a nonfunctional or aromatic residue;
J2 is a nonfunctional residue;
J3 is a hydrophilic residue;
J4 is an acidic residue;
J5 is a nonfunctional residue;
J6 is a basic residue;
J7 is a nonfunctional or aromatic residue;
J8 is a nonfunctional residue;
J11 is a basic or a nonfunctional residue;
J12 is a nonfunctional or aromatic residue;
J16 is a nonfunctional or hydrophilic residue;
J18 is a nonfunctional residue;
J19 is an acidic or basic residue;
J21 is a nonfunctional residue;
JC is J29J30;
J29 is a hydrophilic or nonfunctional residue; and
J30 is a hydrophilic or acidic residue.
26. The composition of matter of claim 25, wherein:
J1 is A, S or Y;
J2 is V;
J3 is S;
J4 is E;
J5 is I;
J6 is Q;
J7 is L or F;
J8 is M or Nle;
J11 is L, R, or K;
J12 is G or W;
J16 is N, S, or A;
J18 is M, Nle, L, or V;
J19 is E or R;
J21 is V, M, or Nle;
J29 is Q or A; and
J30 is D or E.
27. The composition of matter of claim 20, wherein:
JN is J1J2J3J4J5J6;
J1 is a nonfunctional or aromatic residue;
J2 is a nonfunctional residue;
J3 is a hydrophilic residue;
J4 is an acidic residue;
J5 is a nonfunctional residue;
J6 is a basic residue;
J7 is a nonfunctional or aromatic residue;
J8 is a nonfunctional residue;
J11 is a basic or a nonfunctional residue;
J12 is a nonfunctional or aromatic residue;
J16 is a nonfunctional or hydrophilic residue;
J18 is a nonfunctional residue;
J19 is an acidic or basic residue;
J21 is a nonfunctional residue;
JC is J29; and
J29 is a hydrophilic or nonfunctional residue.
28. The composition of matter of claim 27, wherein:
J1 is A, S or Y;
J2 is V;
J3 is S;
J4 is E;
J5 is I;
J6 is Q;
J7 is L or F;
J8 is M or Nle;
J11 is L, R, or K;
J12 is G or W;
J16 is N, S, or A;
J18 is M, Nle, L, or V;
J19 is E or R;
J21 is V, M, or Nle; and
J29 is Q or A.
29. The composition of matter of claim 20, wherein:
JN is J1J2J3J4J5J6;
J1 is a nonfunctional or aromatic residue;
J2 is a nonfunctional residue;
J3 is a hydrophilic residue;
J4 is an acidic residue;
J5 is a nonfunctional residue;
J6 is a basic residue;
J7 is a nonfunctional or aromatic residue;
J8 is a nonfunctional residue;
J11 is a basic or a nonfunctional residue;
J12 is a nonfunctional or aromatic residue;
J16 is a nonfunctional or hydrophilic residue;
J18 is a nonfunctional residue;
J19 is an acidic or basic residue;
J21 is a nonfunctional residue; and
JC is absent.
30. The composition of matter of claim 29, wherein:
J1 is A, S or Y;
J2 is V;
J3 is S;
J4 is E;
J5 is I;
J6 is Q;
J7 is L or F;
J8 is M or Nle;
J11 is L, R, or K;
J12 is G or W;
J16 is N, S, or A;
J18 is M, Nle, L, or V;
J19 is E or R; and
J21 is V, M, or Nle.
31. The composition of matter of claim 20, wherein the PTH/PTHrP modulating domain is selected from Table 1.
32. The composition of matter of claim 1 wherein the PTH/PTHrP modulating domain is of the formula
ONLHO10O11O12KSIO16 (SEQ ID NO: 5) O17LRRRFO23LHHLIOC
wherein:
ON is absent or is YO1O2O3O4O5O6O7, O1O2O3O4O5O6O7, O2O3O4O5O6O7, O3O4O5O6O7, O4O5O6O7, O5O6O7, O6O7, or O7;
O1 is an amino acid residue;
O2 is an amino acid residue;
O3 is an amino acid residue;
O4 is an amino acid residue;
O5 is an amino acid residue;
O6 is an amino acid residue;
O7 is an amino acid residue;
O10 is an amino acid residue;
O11 is an amino acid residue;
O12 is an amino acid residue;
O16 is an amino acid residue;
O17 is an amino acid residue;
O23 is an amino acid residue;
OC is absent or is O29, O29O30, O29O30O31, O29O30O31O32, O29O30O31O32O33, O29O30O31O32O33O34, O29O30O31O32O33O34O35, or O29O30O31O32O33O34O35O36; and
O29 through O36 are each independently amino acid residues.
33. The composition of matter of claim 27, wherein:
ON is O7;
O7 is a nonfunctional residue;
O10 is an acidic or hydrophilic residue;
O11 is a basic or nonfunctional residue;
O12 is an aromatic or nonfunctional residue;
O15 is a hydrophilic or nonfunctional residue;
O16 is a hydrophilic residue;
O17 is an acidic or nonfunctional residue;
O23 is an aromatic residue; and
OC is absent.
34. The composition of matter of claim 23, wherein:
ON is O1O2O3O4O5O6O7;
O1 is a nonfunctional amino acid residue;
O2 is a nonfunctional amino acid residue;
O3 is a hydrophilic amino acid residue;
O4 is an acidic amino acid residue;
O5 is a basic or nonfunctional amino acid residue;
O6 is a hydrophilic amino acid residue;
O7 is a nonfunctional residue;
O10 is an acidic or hydrophilic residue;
O11 is a basic or nonfunctional residue;
O12 is an aromatic or nonfunctional residue;
O15 is a hydrophilic or nonfunctional residue;
O16 is a hydrophilic residue; and
O17 is an acidic or nonfunctional residue; and
O23 is an aromatic residue.
35. The composition of matter of claim 34, wherein:
O1 is A;
O2 is V;
O3 is S;
O4 is E;
O5 is H or I;
O6 is Q;
O7 is L;
O10 is N or D;
O11 is K or L;
O12 is G, F, or W;
O15 is I or S;
O16 is Q or N;
O17 is D or L;
O23 is F or W.
36. The composition of matter of claim 27, wherein the PTH/PTHrP modulating domain is selected from Table 2.
37. The composition of matter of claim 1, wherein the PTH/PTHrP modulating domain has the amino acid sequence of TIP39.
38. The composition of matter of claim 1, comprising a sequence selected from Table 4.
39. A composition of matter, which comprises a peptide selected from SEQ ID NOS: 17,18,19, and 69.
40. A nucleic acid encoding a composition of matter of claim 1.
41. A nucleic acid encoding a composition of matter of claim 7.
42. A nucleic acid encoding a composition of matter of claim 20.
43. A nucleic acid encoding a composition of matter of claim 32.
44. A nucleic acid encoding a composition of matter of claim 39.
45. An expression vector comprising the DNA of claim 40.
46. An expression vector comprising the DNA of claim 41.
47. An expression vector comprising the DNA of claim 42.
48. An expression vector comprising the DNA of claim 43.
49. An expression vector comprising the DNA of claim 44.
50. A host cell comprising the expression vector of claim 45.
51. A host cell comprising the expression vector of claim 46.
52. A host cell comprising the expression vector of claim 47.
53. A host cell comprising the expression vector of claim 48.
54. A host cell comprising the expression vector of claim 49.
55. The cell of claim 50, wherein the cell is an E. coli cell.
56. The cell of claim 51, wherein the cell is an E. coli cell.
57. The cell of claim 52, wherein the cell is an E. coli cell.
58. The cell of claim 53, wherein the cell is an E. coli cell.
59. A process for preparing an antagonist of the PTH/PTHrP receptor, which comprises:
a) selecting at least one peptide that binds to the receptor; and
b) preparing a pharmacologic agent comprising at least one Fc domain covalently linked to at least one amino acid sequence of the selected peptide or peptides.
60. The process of claim 59, wherein the peptide is selected from the SEQ ID NOS: 3,4, or 5.
61. The process of claim 59, wherein the peptide is selected in a process comprising screening of a phage display library, an E. coli display library, a ribosomal library, an RNA-peptide library, or a chemical peptide library.
62. The process of claim 59, wherein the preparation of the pharmacologic agent is carried out by:
a) preparing a gene construct comprising a nucleic acid sequence encoding the selected peptide and a nucleic acid sequence encoding an Fc domain; and
b) expressing the gene construct.
63. The process of claim 59, wherein the gene construct is expressed in an E. coli cell.
64. The process of claim 59, wherein the selection of the peptide is carried out by a process comprising:
a) preparing a gene construct comprising a nucleic acid sequence encoding a first selected peptide and a nucleic acid sequence encoding an Fc domain;
b) conducting a polymerase chain reaction using the gene construct and mutagenic primers, wherein
i) a first mutagenic primer comprises a nucleic acid sequence complementary to a sequence at or near the 5′ end of a coding strand of the gene construct, and
ii) a second mutagenic primer comprises a nucleic acid sequence complementary to the 3′ end of the noncoding strand of the gene construct.
65. A method of treating osteopenia, which comprises administering a PTH agonist and a bone resorption inhibitor, wherein the PTH agonist comprises a composition of matter of claim 1.
66. A method of treating osteopenia, which comprises administering a PTH agonist and a bone resorption inhibitor, wherein the PTH agonist comprises a composition of matter of claim 7.
67. A method of treating osteopenia, which comprises administering a PTH agonist and a bone resorption inhibitor, wherein the PTH agonist comprises a composition of matter of claim 20.
68. A method of treating osteopenia, which comprises administering a PTH agonist and a bone resorption inhibitor, wherein the PTH agonist comprises a composition of matter of claim 32.
69. A method of treating osteopenia, which comprises administering a PTH agonist and a bone resorption inhibitor, wherein the PTH agonist comprises a composition of matter of claim 39.
70. The method of claim 65, wherein the bone resorption inhibitor is selected from OPG, OPG-L antibody, calcitonin, bisphosphonates, estrogens, estrogen receptor modulators, and tibolone.
71. The method of claim 66, wherein the bone resorption inhibitor is selected from OPG, OPG-L antibody, calcitonin, bisphosphonates, estrogens, estrogen receptor modulators, and tibolone.
72. The method of claim 67, wherein the bone resorption inhibitor is selected from OPG, OPG-L antibody, calcitonin, bisphosphonates, estrogens, estrogen receptor modulators, and tibolone.
73. The method of claim 68, wherein the bone resorption inhibitor is selected from OPG, OPG-L antibody, calcitonin, bisphosphonates, estrogens, estrogen receptor modulators, and tibolone.
74. The method of claim 69, wherein the bone resorption inhibitor is selected from OPG, OPG-L antibody, calcitonin, bisphosphonates, estrogens, estrogen receptor modulators, and tibolone.
75. A method of treating osteopenia, which comprises administering a composition of matter of claim 1.
76. A method of treating osteopenia, which comprises administering a composition of matter of claim 7.
77. A method of treating osteopenia, which comprises administering a composition of matter of claim 20.
78. A method of treating osteopenia, which comprises administering a composition of matter of claim 32.
79. A method of treating osteopenia, which comprises administering a composition of matter of claim 39.
US10/839,037 2000-04-27 2004-05-04 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein Abandoned US20040214996A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/839,037 US20040214996A1 (en) 2000-04-27 2004-05-04 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US20005300P 2000-04-27 2000-04-27
US21486000P 2000-06-28 2000-06-28
US26667301P 2001-02-06 2001-02-06
US09/843,221 US6756480B2 (en) 2000-04-27 2001-04-26 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
US10/839,037 US20040214996A1 (en) 2000-04-27 2004-05-04 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/843,221 Division US6756480B2 (en) 2000-04-27 2001-04-26 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein

Publications (1)

Publication Number Publication Date
US20040214996A1 true US20040214996A1 (en) 2004-10-28

Family

ID=27498365

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/843,221 Expired - Fee Related US6756480B2 (en) 2000-04-27 2001-04-26 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
US10/839,037 Abandoned US20040214996A1 (en) 2000-04-27 2004-05-04 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/843,221 Expired - Fee Related US6756480B2 (en) 2000-04-27 2001-04-26 Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein

Country Status (18)

Country Link
US (2) US6756480B2 (en)
EP (1) EP1276767A2 (en)
JP (1) JP2004500838A (en)
KR (1) KR20030004375A (en)
CN (1) CN1439021A (en)
AU (2) AU2001257321B2 (en)
BG (1) BG107242A (en)
BR (1) BR0110391A (en)
CA (1) CA2407493A1 (en)
CZ (1) CZ20023496A3 (en)
EA (1) EA200201121A1 (en)
HK (1) HK1052939A1 (en)
HU (1) HUP0300465A3 (en)
IL (1) IL152384A0 (en)
MX (1) MXPA02010589A (en)
NO (1) NO20025143L (en)
SK (1) SK15202002A3 (en)
WO (1) WO2001081415A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090227498A1 (en) * 2006-10-03 2009-09-10 Radius Health, Inc. Method of drug delivery for bone anabolic protein
US20100029556A1 (en) * 2006-10-03 2010-02-04 Dey Michael J Stable composition comprising a bone anabolic protein, namely a pthrp analogue, and uses thereof
WO2010111617A3 (en) * 2009-03-27 2010-11-18 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
WO2015057836A2 (en) * 2013-10-15 2015-04-23 The Trustees Of Columbia University In The City Of New York Bone anabolic parathyroid hormone and parathyroid hormone related-protein analogs
US10385008B2 (en) 2017-01-05 2019-08-20 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
US11413258B2 (en) 2015-04-29 2022-08-16 Radius Pharmaceuticals, Inc. Methods for treating cancer
USRE49444E1 (en) 2006-10-03 2023-03-07 Radius Health, Inc. Method of treating osteoporosis comprising administration of PTHrP analog
US11643385B2 (en) 2018-07-04 2023-05-09 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCl
US11782041B2 (en) 2017-04-28 2023-10-10 Radius Health, Inc. Abaloparatide formulations and methods of testing, storing, modifying, and using same

Families Citing this family (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6495662B1 (en) * 1998-10-22 2002-12-17 The General Hospital Corporation Bioactive peptides and peptide derivatives of parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHrP)
US7820393B2 (en) * 1999-01-14 2010-10-26 Scantibodies Laboratory, Inc. Methods, kits and antibodies for detecting parathyroid hormone
US6689566B1 (en) * 1999-01-14 2004-02-10 Scantibodies Laboratory, Inc. Methods, kits, and antibodies for detecting parathyroid hormone
WO2001023521A2 (en) * 1999-09-29 2001-04-05 The General Hospital Corporation Polypeptide derivatives of parathyroid hormone (pth)
US20020090646A1 (en) * 2000-05-03 2002-07-11 Amgen Inc. Calcitonin-related molecules
JP4837888B2 (en) * 2001-07-23 2011-12-14 ザ ジェネラル ホスピタル コーポレイション Conformationally restricted parathyroid hormone (PTH) analog
WO2003024486A1 (en) * 2001-09-17 2003-03-27 Chugai Seiyaku Kabushiki Kaisha Remedies for bone loss
US7737260B2 (en) * 2003-11-13 2010-06-15 Hanmi Pharm. Co., Ltd Protein complex using an immunoglobulin fragment and method for the preparation thereof
US7247609B2 (en) 2001-12-18 2007-07-24 Universitat Zurich Growth factor modified protein matrices for tissue engineering
US7015195B2 (en) * 2002-01-10 2006-03-21 Osteotrophin, Llc Treatment of bone disorders with skeletal anabolic drugs
US20050148763A1 (en) * 2002-02-01 2005-07-07 Chugai Seiyaku Kabushiki Kaisha Peg-binding pth or peg-binding pth derivative
US20030191056A1 (en) 2002-04-04 2003-10-09 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
TW200307553A (en) * 2002-05-24 2003-12-16 Akzo Nobel Nv Treatment of post-menopausal complaints in breast cancer patients
US7378495B2 (en) 2002-10-21 2008-05-27 Pevion Biotech, Ltd. PTH-rP related peptide cancer therapeutics
CA2511966A1 (en) * 2002-11-01 2004-07-22 Amgen Inc. Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
US8088734B2 (en) 2003-01-21 2012-01-03 Unigene Laboratories Inc. Oral delivery of peptides
JP4334480B2 (en) * 2003-03-19 2009-09-30 ザ ジェネラル ホスピタル コーポレイション A conformationally constrained parathyroid hormone using an α-helix stabilizer
TWI353991B (en) * 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
AU2016244273B2 (en) * 2003-05-06 2018-10-18 Bioverativ Therapeutics Inc. Immunoglobulin Chimeric Monomer-Dimer Hybrids
WO2004101740A2 (en) 2003-05-06 2004-11-25 Syntonix Pharmaceuticals, Inc. Clotting factor-fc chimeric proteins to treat hemophilia
EP1653985A4 (en) 2003-07-17 2009-08-05 Gen Hospital Corp Conformationally constrained parathyroid hormone (pth) analogs
US7318925B2 (en) * 2003-08-08 2008-01-15 Amgen Fremont, Inc. Methods of use for antibodies against parathyroid hormone
BR0318454A (en) 2003-08-08 2006-09-12 Abgenix Inc parathyroid hormone (pth) targeting antibodies and their uses
US8110665B2 (en) 2003-11-13 2012-02-07 Hanmi Holdings Co., Ltd. Pharmaceutical composition comprising an immunoglobulin FC region as a carrier
CA2562133A1 (en) * 2004-03-19 2005-09-29 Zelos Therapeutics, Inc. Compositions and methods for detecting cyclic analogs of human parathyroid hormone (hpth)
CN101151048A (en) * 2004-05-10 2008-03-26 纳斯泰克制药公司 Compositions and methods for enhanced mucosal delivery of parathyroid hormone
US20060127320A1 (en) * 2004-05-10 2006-06-15 Nastech Pharmaceutical Company Inc. Method of delivering parathyroid hormone to a human
US20060052305A1 (en) * 2004-05-10 2006-03-09 Nastech Pharmaceutical Company Inc. Method of treating osteoporosis using intranasal parathyroid hormone
JP5324781B2 (en) * 2004-06-14 2013-10-23 ビーエーエスエフ、ビューティー、ケア、ソルーションズ、フランス、エスエーエス Cosmetics containing PTH fragments
KR100754667B1 (en) * 2005-04-08 2007-09-03 한미약품 주식회사 Immunoglobulin Fc fragment modified by non-peptide polymer and pharmaceutical composition comprising the same
US8168592B2 (en) * 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
US20070173447A1 (en) * 2005-10-25 2007-07-26 Nastech Pharmaceutical Company Inc. Method for treating osteoporosis by intranasal delivery of teriparatide with an anti-resorptive agent
US7994129B2 (en) * 2005-11-10 2011-08-09 Michigan Technological University Methods of using black bear parathyroid hormone
TW200736277A (en) 2005-11-14 2007-10-01 Amgen Inc RANKL antibody-PTH/PTHrP chimeric molecules
US20080051332A1 (en) * 2005-11-18 2008-02-28 Nastech Pharmaceutical Company Inc. Method of modulating hematopoietic stem cells and treating hematologic diseases using intranasal parathyroid hormone
US8143374B2 (en) * 2006-08-04 2012-03-27 The General Hospital Corporation Polypeptide derivatives of parathyroid hormone (PTH)
CN101522709B (en) * 2006-10-13 2013-03-20 伊莱利利公司 Pegylated PTH as PTH receptor modulators and uses thereof
WO2009131553A2 (en) * 2006-12-29 2009-10-29 Osteogenex Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
JP2008169195A (en) 2007-01-05 2008-07-24 Hanmi Pharmaceutical Co Ltd Insulinotopic peptide drug combo using carrier material
US20090023655A1 (en) * 2007-04-02 2009-01-22 Louis Luttrell Biased Ligands for Receptors Such as the PTH Receptor
WO2008153745A2 (en) 2007-05-22 2008-12-18 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
CA2694667C (en) * 2007-08-01 2018-10-30 The General Hospital Corporation Screening methods using g-protein coupled receptors and related compositions
US20110097318A1 (en) * 2007-08-31 2011-04-28 Amgen Inc. Solid-State Protein Formulation
AU2008304111B2 (en) 2007-09-27 2014-04-24 Amgen Inc. Pharmaceutical formulations
ES2962777T3 (en) 2007-11-15 2024-03-21 Amgen Inc Antioxidant-stabilized aqueous antibody formulation for parenteral administration
EP2227263A2 (en) 2007-12-28 2010-09-15 Kuros Biosurgery AG Pdgf fusion proteins incorporated into fibrin foams
AU2009274738B2 (en) 2008-07-23 2012-12-13 Hanmi Science Co., Ltd. A polypeptide complex comprising non-peptidyl polymer having three functional ends
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
JP2012521197A (en) 2009-03-20 2012-09-13 アムジエン・インコーポレーテツド Carrier immunoglobulin and use thereof
EP2682124B1 (en) * 2009-09-09 2018-02-28 Asahi Kasei Pharma Corporation Pth-containing therapeutic/prophylactic agent for osteoporosis, characterized in that pth is administered once a week at unit dose of 200 units
WO2011050333A1 (en) 2009-10-23 2011-04-28 Amgen Inc. Vial adapter and system
AU2009356227A1 (en) 2009-12-07 2012-06-21 Michigan Technological University Black bear parathyroid hormone and methods of using black bear parathyroid hormone
US9981017B2 (en) 2010-04-02 2018-05-29 Hanmi Science Co., Ltd. Insulin conjugate using an immunoglobulin fragment
AR081066A1 (en) 2010-04-02 2012-06-06 Hanmi Holdings Co Ltd INSULIN CONJUGATE WHERE AN IMMUNOGLOBULIN FRAGMENT IS USED
WO2011143406A2 (en) 2010-05-13 2011-11-17 The General Hospital Corporation Parathyroid hormone analogs and uses thereof
US9061097B2 (en) 2010-06-07 2015-06-23 Amgen Inc. Drug delivery device
KR101382593B1 (en) 2010-07-21 2014-04-10 한미사이언스 주식회사 Novel long-acting glucagon conjugate and pharmaceutical composition comprising the same for the prevention and treatment of obesity
US8993727B2 (en) 2010-09-22 2015-03-31 Amgen Inc. Carrier immunoglobulins and uses thereof
CN103415621A (en) 2011-01-14 2013-11-27 雷德伍德生物科技股份有限公司 Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
MX341790B (en) 2011-03-31 2016-09-02 Amgen Inc Vial adapter and system.
CA2833748C (en) 2011-04-20 2019-07-16 Amgen Inc. Autoinjector apparatus
ES2729993T3 (en) 2011-10-14 2019-11-07 Amgen Inc Injector and assembly procedure
EP2784080B1 (en) * 2011-10-31 2019-12-18 Shimadzu Corporation Peptide-hinge-free flexible antibody-like molecule
KR20130049671A (en) 2011-11-04 2013-05-14 한미사이언스 주식회사 Method for preparation of biological active polypeptide conjugate
DK3391899T3 (en) 2011-12-14 2020-08-24 Univ Arkansas Delivery of therapeutic agents by means of a collagen binding protein
KR101895047B1 (en) 2011-12-30 2018-09-06 한미사이언스 주식회사 A site-specific GLP-2 conjugate using an immunoglobulin fragment
AR090281A1 (en) 2012-03-08 2014-10-29 Hanmi Science Co Ltd IMPROVED PROCESS FOR THE PREPARATION OF A PHYSIOLOGICALLY ACTIVE POLYPEPTIDE COMPLEX
AR091902A1 (en) 2012-07-25 2015-03-11 Hanmi Pharm Ind Co Ltd LIQUID FORMULATION OF A PROLONGED INSULIN CONJUGATE
EP2922590B1 (en) 2012-11-21 2020-02-05 Amgen Inc. Drug delivery device
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
TWI580451B (en) 2013-03-15 2017-05-01 安美基公司 Cassette for an injector and method of using an autoinjector apparatus having an autoinjector and a cassette
LT2976117T (en) 2013-03-22 2021-02-25 Amgen Inc. Injector and method of assembly
US20200354428A9 (en) * 2013-06-23 2020-11-12 Wisconsin Alumni Research Foundation Analogues of parathyroid hormone (1-34) that function as agonists of the parathyroid hormone receptor-1 and display modified activity profiles
AU2014340171B2 (en) 2013-10-24 2019-05-30 Amgen Inc. Injector and method of assembly
WO2015061389A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Drug delivery system with temperature-sensitive control
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
CA3193070A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
JP6817074B2 (en) 2014-06-03 2021-01-20 アムジエン・インコーポレーテツド Controllable drug delivery system and usage
MX2021014323A (en) 2014-10-14 2023-02-02 Amgen Inc Drug injection device with visual and audio indicators.
US11357916B2 (en) 2014-12-19 2022-06-14 Amgen Inc. Drug delivery device with live button or user interface field
EP3233163B1 (en) 2014-12-19 2021-10-13 Amgen Inc. Drug delivery device with proximity sensor
CA3069716C (en) 2015-02-17 2021-11-09 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
EP3981450A1 (en) 2015-02-27 2022-04-13 Amgen, Inc Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
CN106279424A (en) * 2015-05-20 2017-01-04 深圳翰宇药业股份有限公司 A kind of human parathyroid hormone PTH (1-34) fusion protein and application thereof
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
JP7082568B2 (en) 2015-12-09 2022-06-08 アムジエン・インコーポレーテツド Automatic syringe with signal transduction cap
WO2017120178A1 (en) 2016-01-06 2017-07-13 Amgen Inc. Auto-injector with signaling electronics
EP3721922B1 (en) 2016-03-15 2022-05-04 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
US20190119348A1 (en) * 2016-04-01 2019-04-25 The General Hospital Corporation Parathyroid hormone receptor 1 antagonist and inverse agonist polypeptides and methods of their use
CN109071634A (en) 2016-04-26 2018-12-21 R.P.谢勒技术有限责任公司 Antibody coupling matter and its preparation and application
US11541168B2 (en) 2016-04-29 2023-01-03 Amgen Inc. Drug delivery device with messaging label
US11389588B2 (en) 2016-05-02 2022-07-19 Amgen Inc. Syringe adapter and guide for filling an on-body injector
CA3018426A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
EP3465124A1 (en) 2016-06-03 2019-04-10 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
EP3478342A1 (en) 2016-07-01 2019-05-08 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
CN106366176B (en) * 2016-08-30 2020-02-07 杨德鸿 PTH mimetic peptides and uses thereof
US20200261643A1 (en) 2016-10-25 2020-08-20 Amgen Inc. On-body injector
AU2018210301A1 (en) 2017-01-17 2019-08-01 Amgen Inc. Injection devices and related methods of use and assembly
US11624060B2 (en) 2017-02-10 2023-04-11 The Board Of Trustees Of The University Of Arkansas Collagen-binding agent compositions and methods of using the same
EP3582829A1 (en) 2017-02-17 2019-12-25 Amgen Inc. Insertion mechanism for drug delivery device
MX2019009625A (en) 2017-02-17 2019-10-09 Amgen Inc Drug delivery device with sterile fluid flowpath and related method of assembly.
CA3050927A1 (en) 2017-03-06 2018-09-13 Brian Stonecipher Drug delivery device with activation prevention feature
CA3052482A1 (en) 2017-03-07 2018-09-13 Amgen Inc. Needle insertion by overpressure
IL303449B1 (en) 2017-03-09 2024-04-01 Amgen Inc Insertion mechanism for drug delivery device
CN114588404A (en) 2017-03-28 2022-06-07 美国安进公司 Plunger rod and syringe assembly systems and methods
AU2018280054B2 (en) 2017-06-08 2023-07-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
AU2018282077B2 (en) 2017-06-08 2023-11-23 Amgen Inc. Torque driven drug delivery device
AU2018288604B2 (en) 2017-06-22 2023-12-21 Amgen Inc. Device activation impact/shock reduction
MX2019015479A (en) 2017-06-23 2020-02-20 Amgen Inc Electronic drug delivery device comprising a cap activated by a switch assembly.
WO2019014014A1 (en) 2017-07-14 2019-01-17 Amgen Inc. Needle insertion-retraction system having dual torsion spring system
JP2020527376A (en) 2017-07-21 2020-09-10 アムジエン・インコーポレーテツド Gas permeable sealing material and assembly method for drug containers
JP7242562B2 (en) 2017-07-25 2023-03-20 アムジエン・インコーポレーテツド Drug delivery device with container access system and associated method of assembly
EP3658203B1 (en) 2017-07-25 2022-08-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
MA49838A (en) 2017-08-09 2020-06-17 Amgen Inc DRUG DELIVERY SYSTEM WITH CHAMBER HYDRAULIC-PNEUMATIC PRESSURE
US11077246B2 (en) 2017-08-18 2021-08-03 Amgen Inc. Wearable injector with sterile adhesive patch
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
US11759565B2 (en) 2017-10-04 2023-09-19 Amgen Inc. Flow adapter for drug delivery device
EP4257164A3 (en) 2017-10-06 2024-01-17 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
US11464903B2 (en) 2017-10-09 2022-10-11 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
WO2019090086A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
MA50569A (en) 2017-11-06 2020-09-16 Amgen Inc FILLING-FINISHING UNITS AND ASSOCIATED PROCESSES
MA50553A (en) 2017-11-06 2020-09-16 Amgen Inc DRUG ADMINISTRATION DEVICE WITH POSITIONING AND FLOW DETECTION
JP7247174B2 (en) 2017-11-10 2023-03-28 アムジエン・インコーポレーテツド plunger for drug delivery device
AU2018368340B2 (en) 2017-11-16 2024-03-07 Amgen Inc. Door latch mechanism for drug delivery device
MA50903A (en) 2017-11-16 2021-05-12 Amgen Inc SELF-INJECTOR WITH STALL AND END POINT DETECTION
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
MA53375A (en) 2018-07-24 2021-06-02 Amgen Inc ADMINISTRATION DEVICES FOR THE ADMINISTRATION OF MEDICINES
CA3103681A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
US20210228815A1 (en) 2018-07-24 2021-07-29 Amgen Inc. Hybrid drug delivery devices with grip portion
MA53320A (en) 2018-07-31 2021-11-03 Amgen Inc FLUID PATH ASSEMBLY FOR DRUG DELIVERY DEVICE
US20210346601A1 (en) 2018-09-24 2021-11-11 Amgen Inc. Interventional dosing systems and methods
WO2020068476A1 (en) 2018-09-28 2020-04-02 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
AR116679A1 (en) 2018-10-02 2021-06-02 Amgen Inc INJECTION SYSTEMS FOR THE ADMINISTRATION OF DRUGS WITH INTERNAL FORCE TRANSMISSION
US20210338936A1 (en) 2018-10-05 2021-11-04 Amgen Inc. Drug delivery device having dose indicator
EP3866890A1 (en) 2018-10-15 2021-08-25 Amgen Inc. Drug delivery device having damping mechanism
CA3109988A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
EP3873567A1 (en) 2018-11-01 2021-09-08 Amgen Inc. Drug delivery devices with partial needle retraction
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
US11634468B2 (en) * 2019-01-29 2023-04-25 Shire-Nps Pharmaceuticals, Inc. Parathyroid hormone variants
MX2021012557A (en) 2019-04-24 2021-11-12 Amgen Inc Syringe sterilization verification assemblies and methods.
US20220273887A1 (en) 2019-08-23 2022-09-01 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
KR102144688B1 (en) 2020-05-20 2020-08-14 이상진 Waste Styrofoam Recycling Capacity Reduction Device
CA3217207A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4423037A (en) * 1982-05-13 1983-12-27 The General Hospital Corporation Inhibitors of peptide hormone action
US4656250A (en) * 1983-08-05 1987-04-07 Toyo Jozo Kabushiki Kaisha [Nle8, Nle18, Tyr34 or Phe34 ]-h-PTH peptide derivatives
US4968669A (en) * 1988-05-09 1990-11-06 Merck & Co., Inc. Parathyroid hormone antagonists
US5087562A (en) * 1990-04-25 1992-02-11 Merck & Co., Inc. Humoral hypercalcemic factor antagonists with modification at position 13 . . .
US5118667A (en) * 1991-05-03 1992-06-02 Celtrix Pharmaceuticals, Inc. Bone growth factors and inhibitors of bone resorption for promoting bone formation
US5171670A (en) * 1989-05-12 1992-12-15 The General Hospital Corporation Recombinant dna method for production of parathyroid hormone
US5434246A (en) * 1992-03-19 1995-07-18 Takeda Chemical Industries, Ltd. Parathyroid hormone derivatives
US5494806A (en) * 1991-04-05 1996-02-27 The General Hospital Corporation DNA and vectors encoding the parathyroid hormone receptor, transformed cells, and recombinant production of PTHR proteins and peptides
US5529915A (en) * 1993-02-25 1996-06-25 Sterling Winthrop Inc. Lyophilized polyethylene oxide modified protein and polypeptide complexes with cyclodextrin
US5589452A (en) * 1992-07-14 1996-12-31 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5599822A (en) * 1995-06-06 1997-02-04 Eli Lilly And Company Methods for minimizing bone loss
US5599792A (en) * 1992-06-19 1997-02-04 Allelix Biopharmaceuticals Inc. Bone-stimulating, non-vasoactive parathyroid hormone variants
US5670514A (en) * 1995-05-10 1997-09-23 Eli Lilly And Company Compositions for inhibiting bone loss
US5723577A (en) * 1995-07-13 1998-03-03 Biomeasure Inc. Analogs of parathyroid hormone
US5886148A (en) * 1991-04-05 1999-03-23 The General Hospital Corporation Parathyroid hormone receptor
US5955574A (en) * 1995-07-13 1999-09-21 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A. Analogs of parathyroid hormone
US5969095A (en) * 1995-07-13 1999-10-19 Biomeasure, Inc. Analogs of parathyroid hormone
US6147186A (en) * 1996-07-31 2000-11-14 The General Hospital Corporation Parathyroid hormone-related peptide analogs
US6242495B1 (en) * 1997-02-07 2001-06-05 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6417333B1 (en) * 1998-11-25 2002-07-09 The General Hospital Corporation Modified human parathyroid hormone

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU205564B (en) * 1987-01-27 1992-05-28 Laszlo Vadnay Spial spindle for apparatus serving for shelling grains, apparatus for shelling grains and disc for the spiral spindle
US5116964A (en) 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
JPH0532696A (en) * 1990-09-28 1993-02-09 Takeda Chem Ind Ltd Parathyroid hormone derivative
DE69111371T2 (en) 1990-09-28 1996-03-14 Takeda Chemical Industries Ltd Synthetic gene for human parathyroid hormone.
US5556940A (en) * 1994-06-20 1996-09-17 National Research Council Of Canada Parathyroid hormone analogues for the treatment of osteoporosis
US6183974B1 (en) 1997-07-31 2001-02-06 The General Hospital Corporation Screening assays for G protein coupled receptor agonists and antagonists
PL193111B1 (en) 1998-01-29 2007-01-31 Poly Med Inc Absorbable microparticles
DE69907870T2 (en) 1998-01-29 2004-03-04 Kinerton Ltd., Blanchardstown METHOD FOR PRODUCING ABSORBABLE MICROPARTICLES
WO1999046283A1 (en) 1998-03-09 1999-09-16 Zealand Pharmaceuticals A/S Pharmacologically active peptide conjugates having a reduced tendency towards enzymatic hydrolysis
CA2325572A1 (en) 1998-04-15 1999-10-21 Aventis Pharmaceuticals Products Inc. Process for the preparation of resin-bound cyclic peptides
FR2781231B1 (en) 1998-07-17 2002-01-25 Sederma Sa SLIMMING COSMETIC COMPOSITIONS
ATE290214T1 (en) 1998-09-04 2005-03-15 Aventis Pharma Inc LUCIFERASE REPORTER TESTING PROCEDURE FOR PARATHYROID HORMONE COMPOUNDS
AU6510999A (en) 1998-10-07 2000-04-26 Board Of Trustees Of The University Of Arkansas, The Methods of screening for apoptosis-controlling agents for bone anabolic therapies and uses thereof
US6495662B1 (en) 1998-10-22 2002-12-17 The General Hospital Corporation Bioactive peptides and peptide derivatives of parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHrP)
ATE384743T1 (en) 1998-11-25 2008-02-15 Gen Hospital Corp AMINOTERMINAL MODIFIED PARATHYROID HORMONE (PTH) ANALOGUE
WO2000032771A1 (en) * 1998-11-30 2000-06-08 The General Hospital Corporation Pth1r and pth3r receptors, methods and uses thereof
FR2788058B1 (en) 1998-12-30 2002-01-25 Sederma Sa SLIMMING COSMETIC COMPOSITIONS
AU3380200A (en) 1999-02-24 2000-09-14 General Hospital Corporation, The Method for cloning signal transduction intermediates
AU5877300A (en) 1999-06-15 2001-01-02 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Pth2 and pth1 receptor ligands
ECSP003590A (en) 1999-07-31 2002-02-25 Smithkline Beecham Corp CALCIOLITIC COMPOUNDS
WO2001023521A2 (en) 1999-09-29 2001-04-05 The General Hospital Corporation Polypeptide derivatives of parathyroid hormone (pth)
ATE412670T1 (en) 1999-09-29 2008-11-15 Gen Hospital Corp PARATHYROID HORMONE (PTH) POLYPEPTIDE DERIVATIVES

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4423037A (en) * 1982-05-13 1983-12-27 The General Hospital Corporation Inhibitors of peptide hormone action
US4656250A (en) * 1983-08-05 1987-04-07 Toyo Jozo Kabushiki Kaisha [Nle8, Nle18, Tyr34 or Phe34 ]-h-PTH peptide derivatives
US4968669A (en) * 1988-05-09 1990-11-06 Merck & Co., Inc. Parathyroid hormone antagonists
US5171670A (en) * 1989-05-12 1992-12-15 The General Hospital Corporation Recombinant dna method for production of parathyroid hormone
US5087562A (en) * 1990-04-25 1992-02-11 Merck & Co., Inc. Humoral hypercalcemic factor antagonists with modification at position 13 . . .
US5840853A (en) * 1991-04-05 1998-11-24 The General Hospital Corporation Parathyroid hormone receptor and DNA encoding same
US5494806A (en) * 1991-04-05 1996-02-27 The General Hospital Corporation DNA and vectors encoding the parathyroid hormone receptor, transformed cells, and recombinant production of PTHR proteins and peptides
US5886148A (en) * 1991-04-05 1999-03-23 The General Hospital Corporation Parathyroid hormone receptor
US5118667A (en) * 1991-05-03 1992-06-02 Celtrix Pharmaceuticals, Inc. Bone growth factors and inhibitors of bone resorption for promoting bone formation
US5434246A (en) * 1992-03-19 1995-07-18 Takeda Chemical Industries, Ltd. Parathyroid hormone derivatives
US5599792A (en) * 1992-06-19 1997-02-04 Allelix Biopharmaceuticals Inc. Bone-stimulating, non-vasoactive parathyroid hormone variants
US5589452A (en) * 1992-07-14 1996-12-31 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5693616A (en) * 1992-07-14 1997-12-02 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5807823A (en) * 1992-07-14 1998-09-15 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5874086A (en) * 1992-07-14 1999-02-23 Syntex (U.S.A,), Inc. Synthesis and use for the treatment of osteoporosis
US5529915A (en) * 1993-02-25 1996-06-25 Sterling Winthrop Inc. Lyophilized polyethylene oxide modified protein and polypeptide complexes with cyclodextrin
US5670514A (en) * 1995-05-10 1997-09-23 Eli Lilly And Company Compositions for inhibiting bone loss
US5599822A (en) * 1995-06-06 1997-02-04 Eli Lilly And Company Methods for minimizing bone loss
US5723577A (en) * 1995-07-13 1998-03-03 Biomeasure Inc. Analogs of parathyroid hormone
US5955574A (en) * 1995-07-13 1999-09-21 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A. Analogs of parathyroid hormone
US5969095A (en) * 1995-07-13 1999-10-19 Biomeasure, Inc. Analogs of parathyroid hormone
US6147186A (en) * 1996-07-31 2000-11-14 The General Hospital Corporation Parathyroid hormone-related peptide analogs
US6242495B1 (en) * 1997-02-07 2001-06-05 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6417333B1 (en) * 1998-11-25 2002-07-09 The General Hospital Corporation Modified human parathyroid hormone

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8748382B2 (en) 2006-10-03 2014-06-10 Radius Health, Inc. Method of drug delivery for bone anabolic protein
USRE49444E1 (en) 2006-10-03 2023-03-07 Radius Health, Inc. Method of treating osteoporosis comprising administration of PTHrP analog
US20090227498A1 (en) * 2006-10-03 2009-09-10 Radius Health, Inc. Method of drug delivery for bone anabolic protein
US20100029556A1 (en) * 2006-10-03 2010-02-04 Dey Michael J Stable composition comprising a bone anabolic protein, namely a pthrp analogue, and uses thereof
US20110092425A1 (en) * 2006-10-03 2011-04-21 Radius Health, Inc. Method of drug delivery for bone anabolic protein
US8148333B2 (en) 2006-10-03 2012-04-03 Radius Health, Inc. Stable composition comprising a PTHrP analogue
US7803770B2 (en) 2006-10-03 2010-09-28 Radius Health, Inc. Method of treating osteoporosis comprising administration of PTHrP analog
US8563513B2 (en) 2009-03-27 2013-10-22 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
CN102448482A (en) * 2009-03-27 2012-05-09 范安德尔研究所 Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
WO2010111617A3 (en) * 2009-03-27 2010-11-18 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
WO2015057836A3 (en) * 2013-10-15 2015-06-18 The Trustees Of Columbia University In The City Of New York Bone anabolic parathyroid hormone and parathyroid hormone related-protein analogs
WO2015057836A2 (en) * 2013-10-15 2015-04-23 The Trustees Of Columbia University In The City Of New York Bone anabolic parathyroid hormone and parathyroid hormone related-protein analogs
US11413258B2 (en) 2015-04-29 2022-08-16 Radius Pharmaceuticals, Inc. Methods for treating cancer
US11819480B2 (en) 2015-04-29 2023-11-21 Radius Pharmaceuticals, Inc. Methods for treating cancer
US10385008B2 (en) 2017-01-05 2019-08-20 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
US11708318B2 (en) 2017-01-05 2023-07-25 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
US11835506B2 (en) 2017-04-28 2023-12-05 Radius Health, Inc. Abaloparatide formulations and methods of testing, storing, modifying, and using same
US11782041B2 (en) 2017-04-28 2023-10-10 Radius Health, Inc. Abaloparatide formulations and methods of testing, storing, modifying, and using same
US11643385B2 (en) 2018-07-04 2023-05-09 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCl

Also Published As

Publication number Publication date
US20030039654A1 (en) 2003-02-27
MXPA02010589A (en) 2003-03-10
HUP0300465A2 (en) 2003-06-28
IL152384A0 (en) 2003-05-29
JP2004500838A (en) 2004-01-15
WO2001081415A3 (en) 2002-11-28
KR20030004375A (en) 2003-01-14
EP1276767A2 (en) 2003-01-22
BG107242A (en) 2003-06-30
CN1439021A (en) 2003-08-27
NO20025143L (en) 2002-12-23
WO2001081415A2 (en) 2001-11-01
NO20025143D0 (en) 2002-10-25
SK15202002A3 (en) 2003-06-03
US6756480B2 (en) 2004-06-29
BR0110391A (en) 2003-02-25
CA2407493A1 (en) 2001-11-01
HK1052939A1 (en) 2003-10-03
CZ20023496A3 (en) 2003-05-14
EA200201121A1 (en) 2003-06-26
AU2001257321B2 (en) 2007-05-17
WO2001081415A9 (en) 2002-02-14
HUP0300465A3 (en) 2009-11-30
AU5732101A (en) 2001-11-07

Similar Documents

Publication Publication Date Title
US6756480B2 (en) Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
AU2001257321A1 (en) Parathyroid hormone and parathyroid hormone-related protein antagonists
US20050124537A1 (en) Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
US6677136B2 (en) Glucagon antagonists
US6743778B2 (en) Apo-AI/AII peptide derivatives
CA2349406C (en) Compositions and methods for the prevention or treatment of cancer and bone loss associated with cancer
AU2001257173A1 (en) Apo-AI/AII peptide derivatives
AU2002359391A1 (en) Modulators of receptors for parathyrois hormone and parathyroid hormone-related protein
US20020090646A1 (en) Calcitonin-related molecules
AU2005237128B2 (en) Compositions and Methods for the Prevention or Treatment of Cancer and Bone Loss Associated with Cancer
MXPA01004461A (en) Compositions and methods for prevention or treatment of cancer and bone loss associated with cancer

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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