EP1871811A2 - Mimeticorps de la glp-1 humaine, compositions, procedes et utilisations - Google Patents

Mimeticorps de la glp-1 humaine, compositions, procedes et utilisations

Info

Publication number
EP1871811A2
EP1871811A2 EP05858723A EP05858723A EP1871811A2 EP 1871811 A2 EP1871811 A2 EP 1871811A2 EP 05858723 A EP05858723 A EP 05858723A EP 05858723 A EP05858723 A EP 05858723A EP 1871811 A2 EP1871811 A2 EP 1871811A2
Authority
EP
European Patent Office
Prior art keywords
glp
seq
mimetibody
drug
polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP05858723A
Other languages
German (de)
English (en)
Other versions
EP1871811A4 (fr
Inventor
Karyn T. O'neil
Kristen Picha
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.)
Janssen Biotech Inc
Original Assignee
Centocor 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 Centocor Inc filed Critical Centocor Inc
Publication of EP1871811A2 publication Critical patent/EP1871811A2/fr
Publication of EP1871811A4 publication Critical patent/EP1871811A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • 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
    • C07K14/605Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/10Immunoglobulin or domain(s) thereof as scaffolds for inserted non-Ig peptide sequences, e.g. for vaccination purposes

Definitions

  • the present invention relates to mammalian GLP-I mimetibodies, specified portions and variants specific for biologically active proteins, fragment or ligands, GLP-I mimetibody encoding and complementary nucleic acids, host cells, and methods of making and using thereof, including therapeutic formulations, administration and devices.
  • Recombinant proteins are an emerging class of therapeutic agents. Such recombinant therapeutics have engendered advances in protein formulation and chemical modification. Such modifications can potentially enhance the therapeutic utility of therapeutic proteins, such as by increasing half lives (e.g., by blocking their exposure to proteolytic enzymes), enhancing biological activity, or reducing unwanted side effects.
  • One such modification is the use of immunoglobulin fragments fused to receptor proteins, such as enteracept.
  • Therapeutic proteins have also been constructed using the Fc domain to attempt to provide a longer half-life or to incorporate functions such as Fc receptor binding, protein A binding, and complement fixation.
  • Diabetes is a growing epidemic that is estimated to affect over 300 million people by the year 2025 pending an effective pharmaceutical cure. Type 2 diabetes accounts for 90-95% of all cases.
  • Glucagon like peptide-1 is a 37-amino acid peptide secreted from the L- cells of the intestine following an oral glucose challenge.
  • GLP-I (7-37) peptide A subsequent endogenous cleavage between the 6th and 7th position produces the biologically active GLP-I (7-37) peptide.
  • the GLP-I (7-37) peptide sequence can be divided into 2 structural domains. The amino terminal domain of the peptide is involved in signaling while the remainder of the peptide appears to bind to the extracellular loops of the GLP-I receptor in a helical conformation.
  • the active GLP-I binds to the GLP-I receptor on the pancreas and causes an increase in insulin secretion (insulinotropic action).
  • insulin secretion insulin secretion
  • GLP-I has also been shown to inhibit apoptosis and increase proliferation of the ⁇ -cells in the pancreas.
  • GLP-I activity is controlled by blood glucose levels. When blood glucose levels drop to a certain threshold level, GLP-I is not active. Therefore, there is no risk of hypoglycemia associated with treatment involving GLP-I.
  • GLP-I is rapidly inactivated in vivo by the protease dipeptidyl-peptidase IV (DPP-IV). Therefore, the usefulness of therapy involving GLP-I peptides has been limited by their fast clearance and short half-lives.
  • DPP-IV protease dipeptidyl-peptidase IV
  • GLP-I (7-37) has a serum half-life of only 3 to 5 minutes.
  • GLP-I (7-36) amide has a time action of about 50 minutes when administered subcutaneously. Even analogs and derivatives that are resistant to endogenous protease cleavage, do not have half -lives long enough to avoid repeated administrations over a 24 hour period.
  • exenatide is resistant to DPP-IV, yet it still requires twice daily preprandial dosing because of the short half-life and significant variability in in vivo pharmacokinetics.
  • NN2211 another compound currently in clinical trials, is a lipidated GLP-I analogue. It is expected to be dosed once daily.
  • a GLP-I mimetibody may provide a means of delivering the GLP-I peptide in a sustained manner, providing an improvement over GLP-I peptides currently in development. Furthermore, based upon its dimeric structure and its tissue distribution characteristics, a GLP-I mimetibody could have differentiable features with regard to insulin secretion, ⁇ -cell preservation, and food intake.
  • the present invention provides human GLP-I mimetibodies, including modified immunoglobulins, cleavage products and other specified portions and variants thereof, as well as GLP-I mimetibody compositions, encoding or complementary nucleic acids, vectors, host cells, compositions, formulations, devices, transgenic animals, transgenic plants, and methods of making and using thereof, as described and/or enabled herein, in combination with what is known in the art.
  • the present invention also provides at least one isolated GLP-I mimetibody or specified portion or variant as described herein and/or as known in the art.
  • the GLP-I mimetibody can optionally comprise at least one CH3 region directly linked with at least one CH2 region directly linked with at least one portion of at least one hinge region or fragment thereof (H), directly linked with at least one partial variable region (V), directly linked with an optional linker sequence (L), directly linked to at least one GLP- 1 therapeutic peptide (P).
  • a pair of a CH3-CH2-hinge-partial V region sequence-linker-therapeutic peptide sequence the pair optionally linked by association or covalent linkage, such as, but not limited to, at least one Cys-Cys disulfide bond or at least one CH4 or other immunglobulin sequence.
  • a GLP-I mimetibody comprises formula (I): a. (P(n)-L(o)-V(p)-H(q)-CH2(r)-CH3(s))(t),
  • P is at least one bioactive GLP-I peptide, variant or derivative
  • L is at least one linker sequence, which can be a polypeptide that provides structural flexibility by allowing the mimetibody to have alternative orientations and binding properties
  • V is at least one portion of a C-terminus of an immunoglobulin variable region
  • H is at least one portion of an immunoglobulin variable hinge region
  • CH2 is at least a portion of an immunoglobulin CH2 constant region
  • CH3 is at least a portion of an immunoglobulin CH3 constant region
  • n is an integer from 1 to 10
  • o, p, q, r, s, and t can be independently an integer from 0 to 10, mimicing different types of immunoglobulin molecules, e.g., but not limited to IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgM, IgD, IgE, or any subclass thereof, and the like, or any combination thereof.
  • variable region of the antibody sequence can be, but not limited to, at least one portion of at least one of SEQ ID NOS:47-55, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as further described in Figures 1-9 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ ID NOS:47-55, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as further described in Figures 1-9 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ ID NOS:47-55, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as further described in Figures 1-9 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ ID NOS:47-55, or fragment
  • the CH2, CH3 and hinge region can be, but not limited to, at least one portion of at least one of SEQ K) NOS: 56-64, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as as further described in Figures 32-40 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ JX ) NOS:32-40.
  • a GLP-I mimetibody of the present invention mimics at least a portion of an antibody or immnuoglobulin structure or function with its inherent properties and functions, while providing a GLP-I therapeutic peptide and its inherent or acquired in vitro, in vivo or in situ properties or activities.
  • the various portions of the antibody and therapeutic peptide portions of GLP-I mimetibody of the present invention can vary as described herein in combination with what is known in the art.
  • the present invention also provides at least one isolated GLP-I mimetibody or specified portion or variant that has at least one activity, such as, but not limited to known biological activities of at least one bioactive GLP-I peptide or polypeptide corresponding to the P portion of formula (I), as described herein or known in the art.
  • the present invention provides at least one isolated human GLP-I mimetibody comprising at least one polypeptide sequence of SEQ ID NO: 1, or optionally with one or more substitutions, deletions or insertions as described herein or as known in the art.
  • At least one GLP-I mimetibody or specified portion or variant of the invention mimics the binding of at least one GLP-I peptide or polypeptide corresponding to the P portion of the mimetibody in formula (I), to at least one epitope comprising at least 1-3, to the entire amino acid sequence of at least one ligand, e.g., but not limited to, a GLP-I receptor, or fragment thereof, wherein the ligand binds to at least a portion of SEQ ID NO:1, or optionally with one or more substitutions, deletions or insertions as described herein or as known in the art.
  • the at least one GLP- 1 mimetibody can optionally bind GLP-I receptor with an affinity of at least 10 "9 M, at least 10 "10 M, at least 10 "11 M, or at least 10 "12 M.
  • a GLP-I mimetibody can thus be screened for a corresponding activity according to known methods, such as, but not limited to the binding activity towards a receptor or fragment thereof.
  • the present invention further provides at least one anti-idiotype antibody to at least one GLP-I mimetibody of the present invention.
  • the anti-idiotype antibody or fragment specifically binds at least one GLP-I mimetibody of the present invention.
  • the anti-idiotype antibody includes any protein or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule, such as but not limited to at least one complimetarity determing region (CDR) of a heavy or light chain or a ligand binding portion thereof, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or any portion thereof, that competitively binds a GLP-I ligand binding region of at least one GLP-I mimetibody of the present invention.
  • CDR complimetarity determing region
  • Such idiotype antibodies of the invention can include or be derived from any mammal, such as but not limited to a human, a mouse, a rabbit, a rat, a rodent, a primate, and the like.
  • the present invention provides, in one aspect, isolated nucleic acid molecules comprising, complementary, having significant identity or hybridizing to, a polynucleotide encoding at least one GLP-I mimetibody or GLP-I mimetibody antiidiotype antibody, or specified portions or variants thereof, comprising at least one specified sequence, domain, portion or variant thereof.
  • the present invention further provides recombinant vectors comprising at least one of said isolated GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody encoding nucleic acid molecules, host cells containing such nucleic acids and/or recombinant vectors, as well as methods of making and/or using such GLP-I mimetibody or GLP-I mimetibody antiidiotype antibody nucleic acids, vectors and/or host cells.
  • an isolated nucleic acid encoding at least one isolated mammalian GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody; an isolated nucleic acid vector comprising the isolated nucleic acid, and/or a prokaryotic or eukaryotic host cell comprising the isolated nucleic acid.
  • the host cell can optionally be at least one selected from COS-I, COS-7, HEK293, BHK21, CHO, BSC-I, Hep G2, 653, SP2/0, 293, HeLa, myeloma, or lymphoma cells, or any derivative, immortalized or transformed cell thereof.
  • the present invention also provides at least one method for expressing at least one GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody, or specified portion or variant in a host cell, comprising culturing a host cell as described herein and/or as known in the art under conditions wherein at least one GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody, or specified, portion or variant is expressed in detectable and/or recoverable amounts.
  • Also provided is a method for producing at least one GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody comprising translating the GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody encoding nucleic acid under conditions in vitro, in vivo or in situ, such that the GLP-I mimetibody or GLP-I mimetibody anti-idiotype antibody is expressed in detectable or recoverable amounts.
  • the present invention also provides at least one composition
  • a composition comprising (a) an isolated GLP-I mimetibody or specified portion or variant encoding nucleic acid and/or GLP-I mimetibody as described herein; and (b) a suitable carrier or diluent.
  • the carrier or diluent can optionally be pharmaceutically acceptable, according to known methods.
  • the composition can optionally further comprise at least one further compound, protein or composition.
  • compositions comprising at least one isolated human GLP-I mimetibody and at least one pharmaceutically acceptable carrier or diluent.
  • the composition can optionally further comprise an effective amount of at least one compound or protein selected from at least one of a detectable label or reporter, an anti- infective drug, a diabetes or insuling metabolism related drug, a cardiovascular (CV) system drug, a central nervous system (CNS) drug, an autonomic nervous system (ANS) drug, a respiratory tract drug, a gastrointestinal (GI) tract drug, a hormonal drug, a drug for fluid or electrolyte balance, a hematologic drug, an antineoplactic, an immunomodulation drug, an ophthalmic, otic or nasal drug, a topical drug, a nutritional drug, a TNF antagonist, an antirheumatic, a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NTHE), an analgesic, an anesthetic, a sedative,
  • the present invention also provides at least one composition, device and/or method of delivery of a therapeutically or prophylactically effective amount of at least one GLP-I mimetibody or specified portion or variant, according to the present invention.
  • the present invention further provides at least one GLP-I mimetibody method or composition, for administering a therapeutically effective amount to modulate or treat at least one GLP-I related condition in a cell, tissue, organ, animal or patient and/or, prior to, subsequent to, or during a related condition, as known in the art and/or as described herein.
  • the present invention further provides at least one GLP-I mimetibody, specified portion or variant in a method or composition, when administered in a therapeutically effective amount, for modulation, for treating or reducing the symptoms of, at least one metabolic, immune, cardiovascular, infectious, malignant, and/or neurologic disease in a cell, tissue, organ, animal or patient and/or, as needed in many different conditions, such as but not limited to, prior to, subsequent to, or during a related disease or treatment condition, as known in the art.
  • the present invention further provides at least one GLP-I mimetibody, specified portion or variant in a method or composition, when administered in a therapeutically effective amount, for modulation, for treating or reducing the symptoms of at least one of a diabetes or insuling metabolism related disorder, a bone and joint disorder, cardiovascular disoder, a dental or oral disorder, a dermatologic disorder, an ear, nose or throat disorder, an endocrine or metabolic disorder, a gastrointestinal disorder, a gynecologic disorder, a hepatic or biliary disorder, a an obstetric disorder, a hematologic disorder, an immunologic or allergic disorder, an infectious disease, a musculoskeletal disorder, a oncologic disorder, a neurologic disorder, a nutritrional disorder, an opthalmologic disorder, a pediatric disorder, a poisoning disorder, a psychiatric disorder, a renal disorder, a pulmonary disorder, or any other known disorder, (See, e.g., The Merck Manual, a
  • the present invention also provides at least one composition, device and/or method of delivery, for diagnosing GLP-I related conditions, of at least one GLP-I mimetibody, according to the present invention.
  • the present invention further provides at least one GLP-I mimetibody method or composition, for diagnosing at least one GLP-I related condition in a cell, tissue, organ, animal or patient and/or, prior to, subsequent to, or during a related condition, as known in the art and/or as described herein.
  • a method for diagnosing or treating a disease condition in a cell, tissue, organ or animal comprising: (a) contacting or administering a composition comprising an effective amount of at least one isolated human GLP-I mimetibody of the invention with, or to, the cell, tissue, organ or animal.
  • the method can optionally further comprise using an effective amount of 0.001-50 mg/kilogram of the cells, tissue, organ or animal per 0-24 hours, 1-7 days, 1-52 weeks, 1-24 months, 1-30 years or any range or value therein.
  • the method can optionally further comprise using the contacting or the administrating by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal.
  • parenteral subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary,
  • the method can optionally further comprise administering, prior, concurrently or after the (a) contacting or administering, at least one composition comprising an effective amount of at least one compound or protein selected from at least one of a detectable label or reporter, an anti-infective drug, a diabetes or insuling metabolism related drug, a cardiovascular (CV) system drug, a central nervous system (CNS) drug, an autonomic nervous system (ANS) drug, a respiratory tract drug, a gastrointestinal (GI) tract drug, a hormonal drug, a drug for fluid or electrolyte balance, a hematologic drug, an antineoplactic, an immunomodulation drug, an ophthalmic, otic or nasal drug, a topical drug, a nutritional drug, a TNF antagonist, an antirheumatic, a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a local anethetic, a neuromuscular blocker
  • a medical device comprising at least one isolated human GLP- 1 mimetibody of the invention, wherein the device is suitable to contacting or administerting the at least one GLP-I mimetibody by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal.
  • parenteral subcutaneous, intramuscular, intravenous,
  • an article of manufacture for human pharmaceutical or diagnostic use comprising packaging material and a container comprising a solution or a lyophilized form of at least one isolated human GLP-I mimetibody of the present invention.
  • the article of manufacture can optionally comprise having the container as a component of a parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or
  • Figure 1 illustrates the nucleotide and peptide sequences of GLP-I MMB in IgGl scaffold showing important functional domains.
  • FIGS 2A-2C illustrate FACS binding assays of GLP-I MMB.
  • Figure 2A shows that GLP-I MMB binds to HEK293 cells over-expressing the GLP-IR. Grey area: GLP-I MMB but no secondary; grey line: secondary only; dotted line, negative control MMB and secondary; black line: GLP-I MMB and secondary.
  • Figure 2B shows that the GLP-I MMB does not bind to the control HEK293 cells. Grey area: GLP-I MMB but no secondary; black line: secondary only; grey line: GLP-I MMB and secondary.
  • Figure 2C shows that a GLP-I peptide analogue (A2S) is able to compete with GLP- 1 MMB for binding to HEK293 cells over-expressing the GLP- 1 R.
  • Grey area GLP-I MMB but no secondary; black line: GLP-I MMB and secondary; break line: GLP-I MMB, 0.2 nM competitor, secondary; dotted line: GLP-I MMB, 20 nM competitor, secondary; grey line: GLP-I MMB, 100 nM competitor, secondary).
  • Figures 3A-3E illustrate cAMP assays of GLP-I MMB.
  • Figure 3 A wt GLP-I MMB in IgGl scaffold
  • Figure 3B GLP-I peptide
  • Figure 3C GLP-I (A2G) MMB in IgG4 (Ala/Ala, Ser -> Pro) scaffold
  • Figure 3D GLP-I (A2S) MMB in IgG4 (Ala/Ala, Ser -> Pro) scaffold
  • Figure 3E wt GLP-I MMB in IgG4 (Ala/Ala, Ser -> Pro) scaffold.
  • Figure 4 illustrates the resistance of GLP-I MMB to DPP-IV cleavage.
  • Figure 5 shows the stability of GLP-I MMB in serum.
  • Figure 6 demonstrates that GLP-I MMBs cause insulin secretion in RlNm cells.
  • Figure 6A shows that GLP-I (7-36) peptide and exendin-4 peptide stimulates insulin release in RINm cells.
  • Figure 6B shows that GLP-I (A2S) MMB in either IgGl or IgG4 (Ala/Ala, Ser -> Pro) scaffold, or GLP-I (A2G) MMB in IgG4 (Ala/Ala, Ser -> Pro) scaffold are active in stimulating insulin secretion in RINm cells.
  • Figure 7 demonstrates that GLP-I MMB lowers glucose (Figure 7A) in a dose-dependent manner (figure 7B).
  • Figure 8 shows the pharmacokinetic profile of four GLP-I MMBs (A2G, A2S, Ex-cap and wild-type) in cynomolgus monkey.
  • Figure 9 shows the effects of GLP-I MMB during an oral glucose tolerance test in diabetic mice.
  • Figure 10 shows the effects of GLP-I MMB on fasting blood glucose during chronic dosing to diabetic mice.
  • Figure 11 shows the effects of GLP-I MMB on oral glucose tolerance test after chronic dosing to diabetic mice.
  • Figure 12 shows the effects of GLP-I MMB on reducing HbAIc after chronic dosing to diabetic mice.
  • Figure 13 shows the effects of GLP-I MMB on blood glucose (Figure 13A) and insulin (Figure 13B) levels in an oral glucose tolerance test in normal cynomolgus monkeys.
  • Figure 14 shows the effects of GLP-I MMB on insulin staining in islets of diabetic mice after a single dose.
  • Figure 15 demonstrates that GLP-I MMB delays gastric emptying in normal dogs.
  • Figure 16 demonstrates that GLP-I MMB lowers blood glucose following an oral glucose tolerance test in diet induced obese mice.
  • Figure 17 demonstrates that GLP-I MMB lowers blood glucose (Figure 17A) and lowers insulin level (Figure 17B) in an intraperitoneal glucose tolerance test in diabetic mice.
  • Figure 18 demonstrates that GLP-I mimetibody inhibits cytokine-induced apoptosis in a dose-dependent manner. The percent of apoptosis relative to the untreated control is plotted versus the concentration of GLP-I MMB.
  • FIG. 19 demonstrates that GLP-I mimetibody increases glucose-dependent insulin secretion in INS-IE cells.
  • A. The bar graph shows the amount of insulin secreted at each concentration of GLP-I MMB in the presence of 5.5 mM glucose. In addition, the insulin secreted at 3 and 7.5 mM glucose only is plotted.
  • Figure 20 demonstrates that GLP-I mimetibody increases glucose-dependent insulin secretion in rat (Figure 20A) and human islets (Figure 20B).
  • Figure 21 shows the effects of GLP-I MMB treatment on the blood glucose levels of normal C57/BLK6 mice.
  • Figure 22 shows the effects of GLP-I MMB treatment on the blood glucose levels (Figure 22A) and glucose tolerance (Figure 22B) in STZ treated diabetic nude mice transplanted with a marginal Mass of human islets.
  • Figure 22 A blood glucose was plotted against time (days) for control mice treated with PBS (X) and GLP-I MMB treated mice ( ⁇ ) receiving daily IP injections.
  • a marginal mass of human islets (50 IEQ/g) was transplanted on day 0 and blood glucose in the animals was monitored thereafter. The arrow indicates when the transplanted islets were removed, resulting in the prompt return to the diabetic state.
  • FIG 22B blood glucose plotted against time (minutes) for control mice treated with PBS (X) and GLP-I MMB (0.5 mg/kg) ( ⁇ ).
  • the rVGTT was done after the mice received daily IP injections for >30 days. Animals were fasted overnight followed by an IV glucose injection (1.5 g/kg). The blood glucose of the mice was monitored and plotted against time post injection. The insert represents the area under the curve for the control and GLP-I MMB treated animals.
  • Figure 23 shows the effects of GLP-I MMB on glucose-induced insulin secretion in non-human primate (NHP) islets.
  • NHS non-human primate
  • Figure 24 shows the effects of GLP-I MMB on the exogenous insulin requirements (Figure 24A) and the Hemoglobin AIc (HgbAlc) ( Figure 24B) of STZ diabetic NHPs transplanted with a marginal mass.
  • the present invention provides isolated, recombinant and/or synthetic mimetibodies or specified portions or variants, as well as compositions and encoding nucleic acid molecules comprising at least one polynucleotide encoding at least one GLP-I mimetibody.
  • Such mimetibodies or specified portions or variants of the present invention comprise specific GLP-I mimetibody sequences, domains, fragments and specified variants thereof, and methods of making and using said nucleic acids and mimetibodies or specified portions or variants, including therapeutic compositions, methods and devices.
  • the present invention also provides at least one isolated GLP-I mimetibody or specified portion or variant as described herein and/or as known in the art.
  • the GLP-I mimetibody can optionally comprise at least one CH3 region directly linked with at least one CH2 region directly linked with at least one hinge region or fragment thereof (H), directly linked with at least one partial variable region (V), directly linked with an optional linker sequence (L), directly linked to at least one GLP-I therapeutic peptide (P)-
  • Li a preferred embodiment a GLP-I mimetibody comprises formula (I): ((P(n)-L(o)-V(p)-H(q)-CH2(r)-CH3(s))(t),
  • P is at least one bioactive GLP-I polypeptide
  • L is at least one linker sequence, which can be a polypeptide that provides structural flexablity by allowing the mimietibody to have alternative orientations and binding properties
  • V is at least one portion of a C-terminus of an immunoglobulin variable region
  • H is at least one portion of an immunoglobulin variable hinge region
  • CH2 is at least a portion of an immunoglobulin CH2 constant region
  • CH3 is at least a portion of an immunoglobulin CH3 constant region
  • m, n, o, p, q, r, s and t can be independently an integer between and including 0 and 10, mimicing different types of immunoglobulin molecules, e.g., but not limited to IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgM, IgD, IgE, or any subclass thereof, and the like, or any combination thereof.
  • a GLP-I mimetibody of the present invention mimics an antibody structure with its inherent properties and functions, while providing a therapeutic peptide and its inherent or acquired in vitro, in vivo or in situ properties or activities.
  • the monomer CH3-CH2-hinge-partial J sequence- linker-therapeutic peptide can be linked to other monomers by association or covalent linkage, such as, but not limited to, a Cys-Cys disulfide bond.
  • the various portions of the antibody and the GLP-I therapeutic peptide portions of at least one GLP-I mimetibody of the present invention can vary as described herein in combinatoin with what is known in the art.
  • the portion of CH3-CH2-hinge may be extensively modified to form a variant in accordance with this invention, provided binding to the salvage receptor is maintained.
  • one may remove one or more native sites 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.
  • a variant of CH3-CH2-hinge may lack one or more native sites or residues that affect or are involved in (1) disulfide bond formation, (2) incompatibility with a selected host cell, (3) 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).
  • Exemplary CH3-CH2-hinge variants include molecules and sequences in which: 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.
  • cysteine residues may be deleted or substituted with other amino acids (e.g., alanyl, seryl). Even when cysteine residues are removed, the single chain CH3-CH2-hinge domains can still form a dimeric CH3-CH2-hinge domain that is held together non-covalently; 2.
  • the CH3-CH2-hinge region is modified to make it more compatible with a selected host cell. For example, when the molecule is expressed recombinantly in a bacterial cell such as E. coli, one may remove the PA sequence in the hinge, which may be recognized by a digestive enzyme in E. coli such as proline iminopeptidase; 3.
  • a portion of the hinge region is deleted or substituted with other amino acids to prevent heterogeneity when expressed in a selected host cell; 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); 5.
  • Sites involved in interaction with complement, such as the CIq binding site, are removed. Complement recruitment may not be advantageous for the molecules of this invention and so may be avoided with such a variant; 6.
  • Sites are removed that affect binding to Fc receptors other than a salvage receptor.
  • the CH3-CH2-hinge region 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; 7.
  • the 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 IgGl. These sites, as well, are not required for the fusion molecules of the present invention and so may be removed.
  • Linker polypeptide provides structural flexibility by allowing the mimetibody to have alternative orientations and binding properties. When present, its chemical structure is not critical.
  • 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, serine, 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 poly(Gly- Ser), polyglycines (particularly (GIy) 4 , (GIy) 5 ), poly(Gly-Ala), and polyalanines.
  • Other specific examples of linkers are: (GIy) 3 LyS(GIy) 4 (SEQ ID NO:65), (Gly) 3 AsnGlySer(Gly) 2 (SEQ ID NO:66),
  • (Gly) 3 Lys(Gly) 4 means GIy- Gly-Gly-Lys-Gly-Gly-Gly-Gly. Combinations of GIy 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, NH2, phenyl, etc.
  • An exemplary non-peptide linker is a PEG linker which 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.
  • GLP-I peptide can be at least one GLP-I peptide, GLP-I fragment, GLP-I homolog, GLP-I analog, or GLP-I derivative.
  • a GLP-I peptide has from about twenty-five to about forty-five naturally occurring or non-naturally occurring amino acids that have sufficient homology to native GLP-I (7-37) such that they exhibit insulinotropic activity by binding to the GLP-I receptor on ⁇ -cells in the pancreas.
  • GLP-I (7-37) has the amino acid sequence of SEQ ID NO: 15 :
  • a GLP-I fragment is a polypeptide obtained after truncation of one or more amino acids from the N-terminus and/or C-terminus of GLP-I (7-37) or an analog or derivative thereof.
  • a GLP-I homolog is a peptide in which one or more amino acids have been added to the N-terminus and/or C-terminus of GLP-I (7-37), or fragments or analogs thereof.
  • a GLP-I analog is a peptide in which one or more amino acids of GLP-I (7-37) have been modified and/or substituted.
  • a GLP-I analog has sufficient homology to GLP-I (7-37) or a fragment of GLP-I (7-37) such that the analog has insulinotropic activity.
  • a GLP-I derivative is defined as a molecule having the amino acid sequence of a GLP-I peptide, a GLP-I homolog or a GLP-I analog, but additionally having chemical modification of one or more of its amino acid side groups, ⁇ -carbon atoms, terminal amino group, or terminal carboxylic acid group.
  • GLP-I fragments, analogs and derivatives are known in the art and any of these analogs and derivatives can also be part of the GLP-I mimetibody of the present invention.
  • Some GLP-I analogs and GLP-I fragments known in the art are disclosed in U.S. Pat. Nos. 5,118,666, 5,977,071, and 5,545,618, and Adelhorst, et al., J. Biol. Chem. 269:6275 (1994).
  • Examples include, but not limited to, GLP-I (7-34), GLP- 1 (7-35), GLP-I (7-36), Gln9-GLP- 1(7-37), D-Gln9-GLP- 1(7-37), Thrl6-Lysl8-GLP-1 (7-37), and Lysl8-GLP-1 (7-37).
  • a "GLP-I mimetibody,” “GLP-I mimetibody portion,” or “GLP-I mimetibody fragment” and/or “GLP-I mimetibody variant” and the like has, mimics or simulates at least one biological activity, such as but not limited to ligand binding, in vitro, in situ and/or preferably in vivo, of at least one GLP-I peptide, variant or derivative, such as but not limited to at least one of SEQ ID NO: 1.
  • a suitable GLP- 1 mimetibody, specified portion, or variant can also modulate, increase, modify, activate, at least one GLP-I receptor signaling or other measurable or detectable activity.
  • GLP-I mimetibodies useful in the methods and compositions of the present invention are characterized by suitable affinity binding to protein ligands, for example, GLP-I receptors, and optionally and preferably having low toxicity.
  • a GLP-I mimetibody where the individual components, such as the portion of variable region, constant region (without a CHl portion) and framework, or any portion thereof (e.g., a portion of the J, D or V rgions of the variable heavy or light chain; at least a portion of at least one hinge region, the constant heavy chain or light chain, and the like) individually and/or collectively optionally and preferably possess low immunogenicity, is useful in the present invention.
  • the mimetibodies that can be used in the invention are optionally characterized by their ability to treat patients for extended periods with good to excellent alleviation of symptoms and low toxicity. Low immunogenicity and/or high affinity, as well as other undefined properties, may contribute to the therapeutic results achieved.
  • Low immunogenicity is defined herein as raising significant HAMA, HACA or HAHA responses in less than about 75%, or preferably less than about 50, 45, 40, 35, 30, 35, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, and/or 1% of the patients treated and/or raising low titres in the patient treated (less than about 300, preferably less than about 100 measured with a double antigen enzyme immunoassay) (see, e.g., Elliott et al., Lancet 344:1125-1121 (1994)).
  • the isolated nucleic acids of the present invention can be used for production of at least one GLP-I mimetibody, fragment or specified variant thereof, which can be used to effect in an cell, tissue, organ or animal (including mammals and humans), to modulate, treat, alleviate, help prevent the incidence of, or reduce the symptoms of, at least one protein related condition, selected from, but not limited to, at least one of a diabetes related disorder, an insulin metabolism related disorder, an immune disorder or disease, a cardiovascular disorder or disease, an infectious, malignant, and/or neurologic disorder or disease, as well as other known or specified protein related conditions.
  • Such a method can comprise administering an effective amount of a composition or a pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment, alleviation, prevention, or reduction in symptoms, effects or mechanisms.
  • the effective amount can comprise an amount of about 0.0001 to 500 mg/kg per single or multiple administration, or to achieve a serum concentration of 0.01-5000 ⁇ g/ml serum concentration per single or multiple adminstration, or any effective range or value therein, as done and determined using known methods, as described herein or known in the relevant arts.
  • the GLP-I mimetibody can optionally comprise at least one CH3 region directly linked with at least one CH2 region directly linked with at least one portion of at lesat one hinge region fragment (H), such as comprising at least one core hinge region, directly linked with at least one partial variable region (V), directly linked with an optional linker sequence (L), directly linked to at least one GLP-I therapeutic peptide (P).
  • H hinge region fragment
  • V partial variable region
  • L optional linker sequence
  • P GLP-I therapeutic peptide
  • a pair of a CH3-CH2-H-V-L-P can be linked by association or covalent linkage, such as, but not limited to, a Cys-Cys disulfide bond.
  • a GLP-I mimetibody of the present invention mimics an antibody structure with its inherent properties and functions, while providing a. therapeutic peptide and its inherent or acquired in vitro, in vivo or in situ properties or activities.
  • the various portions of the antibody and therapeutic peptide portions of at least one GLP-I mimetibody of the present invention can vary as described herein in combinatoin with what is known in the art.
  • Mimetibodies of the present invention thus provide at least one suitable property as compared to known proteins, such as, but not limited to, at least one of increased half-life, increased activity, more specific activity, increased avidity, increased or descrease off rate, a selected or more suitable subset of activities, less immieuxicity, increased quality or duration of at least one desired therapeutic effect, less side effects, and the like.
  • Fragments of mimetibodies according to Formula (I) can be produced by enzymatic cleavage, synthetic or recombinant techniques, as known in the art and/or as described herein.
  • Mimetibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site.
  • the various portions of mimetibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques.
  • a nucleic acid encoding at least one of the constant regions of a human antibody chain can be expressed to produce a contiguous protein for use in mimetibodies of the present invention. See, e.g., Ladner et ah, U.S. Patent No. 4,946,778 and Bird, R.E. et al, Science, 242: 423-426 (1988), regarding single chain antibodies.
  • human mimetibody refers to an antibody in which substantially every part of the protein (e.g., GLP-I peptide, C H domains (e.g., Q H 2, C H 3), hinge, V) is expected to be substantially non-immunogenic in humans with only minor sequence changes or variations. Such changes or variations optionally and preferably retain or reduce the immunogenicity in humans relative to non-modified human antibodies, or mimetibodies of the prsent invention.
  • a human antibody and corresponding GLP-I mimetibody of the present invention is distinct from a chimeric or humanized antibody. It is pointed out that the GLP-I mimetibody can be produced by a non-human animal or cell that is capable of expressing human immunoglobulins (e.g., heavy chain and/or light chain) genes.
  • Human mimetibodies that are specific for at least one protein ligand thereof can be designed against an appropriate ligand, such as an isolated GLP-I receptor, or a portion thereof (including synthetic molecules, such as synthetic peptides). Preparation of such mimetibodies are performed using known techniques to identify and characterize ligand binding regions or sequences of at least one protein or portion thereof.
  • At least one GLP-I mimetibody or specified portion or variant of the present invention is produced by at least one cell line, mixed cell line, immortalized cell or clonal population of immortalized and/or cultured cells.
  • Immortalized protein producing cells can be produced using suitable methods.
  • the at least one GLP-I mimetibody or specified portion or variant is generated by providing nucleic acid or vectors comprising DNA derived or having a substantially similar sequence to, at least one human immunoglobulin locus that is functionally rearranged, or which can undergo functional rearrangement, and which further comprises a mimetibody structure as described herein, e.g., but not limited to Formula (I), wherein portions of C- terminal variable regions can be used for V, hinge regions for H, CH2 for CH2 and CH3 for CH3, as known in the art.
  • Formula (I) wherein portions of C- terminal variable regions can be used for V, hinge regions for H, CH2 for CH2 and CH3 for CH3, as known in the art.
  • the term "functionally rearranged,” as used herein refers to a segment of nucleic acid from an immunoglobulin locus that has undergone V(D)J recombination, thereby producing an immunoglobulin gene that encodes an immunoglobulin chain (e.g., heavy chain), or any portion thereof.
  • a functionally rearranged immunoglobulin gene can be directly or indirectly identified using suitable methods, such as, for example, nucleotide sequencing, hybridization (e.g., Southern blotting, Northern blotting) using probes that can anneal to coding joints between gene segments or enzymatic amplification of immunoglobulin genes (e.g., polymerase chain reaction) with primers that can anneal to coding joints between gene segments.
  • Mimetibodies, specified portions and variants of the present invention can also be prepared using at least one GLP-I mimetibody or specified portion or variant encoding nucleic acid to provide transgenic animals or mammals, such as goats, cows, horses, sheep, and the like, that produce such mimetibodies or specified portions or variants in their milk. Such animals can be provided using known methods as applied for antibody encoding sequences. See, e.g., but not limited to, US patent nos.
  • Mimetibodies, specified portions and variants of the present invention can additionally be prepared using at least one GLP-I mimetibody or specified portion or variant encoding nucleic acid to provide transgenic plants and cultured plant cells (e.g., but not limited to tobacco and maize) that produce such mimetibodies, specified portions or variants in the plant parts or in cells cultured therefrom.
  • transgenic tobacco leaves expressing recombinant proteins have been successfully used to provide large amounts of recombinant proteins, e.g., using an inducible promoter. See, e.g., Cramer et al., Curr. Top. Microbol. Immunol. 240:95-118 (1999) and references cited therein.
  • transgenic maize or corn have been used to express mammalian proteins at commercial production levels, with biological activities equivalent to those produced in other recombinant systems or purified from natural sources. See, e.g., Hood et al., Adv. Exp. Med. Biol. 464:127-147 (1999) and references cited therein.
  • Antibodies have also been produced in large amounts from transgenic plant seeds including antibody fragments, such as single chain mimetibodies (scFv's), including tobacco seeds and potato tubers. See, e.g., Conrad et al., Plant MoI. Biol. 38:101-109 (1998) and references cited therein.
  • mimetibodies, specified portions and variants of the present invention can also be produced using transgenic plants, according to know methods. See also, e.g., Fischer et al., Biotechnol. Appl. Biochem. 30:99-108 (Oct., 1999), Ma et al., Trends Biotechnol. 13:522-7 (1995); Ma et al., Plant Physiol. 109:341-6 (1995); Whitelam et al., Biochem. Soc. Trans. 22:940-944 (1994); and references cited therein. The above references are entirely incorporated herein by reference.
  • the mimetibodies of the invention can bind human protein ligands with a wide range of affinities (K D ).
  • at least one human GLP-I mimetibody of the present invention can optionally bind at least one protein ligand with high affinity.
  • At least one GLP-I mimetibody of the present invention can bind at least one protein ligand with a K D equal to or less than about 10 "7 M or, more preferably, with a K D equal to of less than about 0.1-9.9 (or any range or value therein) x 10 '7 , 10 "8 , 10 "9 , 10 "10 , 10 "11 , 10 "12 , or 10 '13 M, or any range or value therein.
  • the affinity or avidity of a GLP-I mimetibody for at least one protein ligand can be determined experimentally using any suitable method, e.g., as used for determing antibody-antigen binding affinity or avidity.
  • any suitable method e.g., as used for determing antibody-antigen binding affinity or avidity.
  • the measured affinity of a particular GLP-I mimetibody-ligand interaction can vary if measured under different conditions (e.g., salt concentration, pH).
  • affinity and other ligand-binding parameters e.g., K D , K 3 , K d
  • K D , K 3 , K d affinity and other ligand-binding parameters
  • nucleic Acid Molecules Using the information provided herein, such as the nucleotide sequences encoding at least 90-100% of the contiguous amino acids of at least one of SEQ ID NOS: 1 and 6, as well as at least one portion of an antibody, wherein the above sequences are inserted as the P sequence of Formula (I) to provide a GLP-I mimetibody of the present invention, further comprising specified fragments, variants or consensus sequences thereof, or a deposited vector comprising at least one of these sequences, a nucleic acid molecule of the present invention encoding at least one GLP-I mimetibody or specified portion or variant can be obtained using methods described herein or as known in the art.
  • Nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, hnRNA, tRNA or any other form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or produced synthetically, or any combination thereof.
  • the DNA can be triple-stranded, double-stranded or single- stranded, or any combination thereof. Any portion of at least one strand of the DNA or RNA can be the coding strand, also known as the sense strand, or it can be the non- coding strand, also referred to as the anti-sense strand.
  • Isolated nucleic acid molecules of the present invention can include nucleic acid molecules comprising an open reading frame (ORF), optionally with one or more introns, nucleic acid molecules comprising the coding sequence for a GLP-I mimetibody or specified portion or variant; and nucleic acid molecules which comprise a nucleotide sequence substantially different from those described above but which, due to the degeneracy of the genetic code, still encode at least one GLP-I mimetibody as described herein and/or as known in the art.
  • ORF open reading frame
  • nucleic acid molecules comprising the coding sequence for a GLP-I mimetibody or specified portion or variant
  • the genetic code is well known in the art.
  • nucleic acid variants that code for specific GLP-I mimetibody or specified portion or variants of the present invention. See, e.g., Ausubel, et al., supra, and such nucleic acid variants are included in the present invention.
  • nucleic acid molecules of the present invention which comprise a nucleic acid encoding a GLP-I mimetibody or specified portion or variant can include, but are not limited to, those encoding the amino acid sequence of a GLP-I mimetibody fragment, by itself; the coding sequence for the entire GLP-I mimetibody or a portion thereof; the coding sequence for a GLP-I mimetibody, fragment or portion, as well as additional sequences, such as the coding sequence of at least one signal leader or fusion peptide, with or without the aforementioned additional coding sequences, such as at least one intron, together with additional, non-coding sequences, including but not limited to, non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals (for example - ribosome binding and stability of mRNA); an additional coding sequence that codes for additional
  • sequence encoding a GLP-I mimetibody. or specified portion or variant can be fused to a marker sequence, such as a sequence encoding a peptide that facilitates purification of the fused GLP- 1 mimetibody or specified portion or variant comprising a GLP-I mimetibody fragment or portion.
  • polynucleotides which Selectively Hybridize to a Polynucleotide as Described Herein.
  • the present invention provides isolated nucleic acids that hybridize under selective hybridization conditions to a polynucleotide disclosed herein, or others disclosed herein, including specified variants or portions thereof.
  • the polynucleotides of this embodiment can be used for isolating, detecting, and/or quantifying nucleic acids comprising such polynucleotides.
  • Low or moderate stringency hybridization conditions are typically, but not exclusively, employed with sequences having a reduced sequence identity relative to complementary sequences. Moderate and high stringency conditions can optionally be employed for sequences of greater identity. Low stringency conditions allow selective hybridization of sequences having about 40-99% sequence identity and can be employed to identify orthologous or paralogous sequences.
  • polynucleotides of this invention will encode at least a portion of a GLP-I mimetibody or specified portion or variant encoded by the polynucleotides described herein.
  • the polynucleotides of this invention embrace nucleic acid sequences that can be employed for selective hybridization to a polynucleotide encoding a GLP-I mimetibody or specified portion or variant of the present invention. See, e.g., Ausubel, supra; Colligan, supra, each entirely incorporated herein by reference.
  • Construction of Nucleic Acids The isolated nucleic acids of the present invention can be made using (a) recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well-known in the art.
  • the nucleic acids can conveniently comprise sequences in addition to a polynucleotide of the present invention.
  • a multi-cloning site comprising one or more endonuclease restriction sites can be inserted into the nucleic acid to aid in isolation of the polynucleotide.
  • translatable sequences can be inserted to aid in the isolation of the translated polynucleotide of the present invention.
  • a hexa- histidine marker sequence provides a convenient means to purify the proteins of the present invention.
  • the nucleic acid of the present invention - excluding the coding sequence - is optionally a vector, adapter, or linker for cloning and/or expression of a polynucleotide of the present invention.
  • Additional sequences can be added to such cloning and/or expression sequences to optimize their function in cloning and/or expression, to aid in isolation of the polynucleotide, or to improve the introduction of the polynucleotide into a cell.
  • Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art. See, e.g., Ausubel, supra; or Sambrook, supra.
  • RNA, cDNA, genomic DNA, or any combination thereof can be obtained from biological sources using any number of cloning methodologies known to those of skill in the art.
  • oligonucleotide probes that selectively hybridize, under suitable stringency conditions, to the polynucleotides of the present invention are used to identify the desired sequence in a cDNA or genomic DNA library.
  • the isolation of RNA, and construction of cDNA and genomic libraries, is well known to those of ordinary skill in the art. (See, e.g., Ausubel, supra; or Sambrook, supra).
  • the isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by known methods (see, e.g., Ausubel, et al., supra). Chemical synthesis generally produces a single-stranded oligonucleotide, which can be converted into double- stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template.
  • a complementary sequence or by polymerization with a DNA polymerase using the single strand as a template.
  • One of skill in the art will recognize that while chemical synthesis of DNA can be limited to sequences of about 100 or more bases, longer sequences can be obtained by the ligation of shorter sequences.
  • the present invention further provides recombinant expression cassettes comprising a nucleic acid of the present invention.
  • a nucleic acid sequence of the present invention for example a cDNA or a genomic sequence encoding a GLP-I mimetibody or specified portion or variant of the present invention, can be used to construct a recombinant expression cassette that can be introduced into at least one desired host cell.
  • a recombinant expression cassette will typically comprise a polynucleotide of the present invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell. Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the present invention.
  • isolated nucleic acids that serve as promoter, enhancer, or other elements can be introduced in the appropriate position (upstream, downstream or in intron) of a non-heterologous form of a polynucleotide of the present invention so as to up or down regulate expression of a polynucleotide of the present invention.
  • endogenous promoters can be altered in vivo or in vitro by mutation, deletion and/or substitution, as known in the art.
  • a polynucleotide of the present invention can be expressed in either sense or anti-sense orientation as desired. It will be appreciated that control of gene expression in either sense or anti-sense orientation can have a direct impact on the observable characteristics. Another method of suppression is sense suppression.
  • the present invention also relates to vectors that include isolated nucleic acid molecules of the present invention, host cells that are genetically engineered with the recombinant vectors, and the production of at least one GLP-I mimetibody or specified portion or variant by recombinant techniques, as is well known in the art. See, e.g., Sambrook, et al., supra; Ausubel, et al, supra, each entirely incorporated herein by reference.
  • the polynucleotides can optionally be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced into a cell using suitable known methods, such as electroporation and the like, other known methods include the use of the vector as a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it can be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the DNA insert should be operatively linked to an appropriate promoter.
  • the expression constructs will further contain sites optionally for at least one of transcription initiation, termination and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the mature transcripts expressed by the constructs will preferably include a translation initiating at the beginning and a termination codon (e.g., UAA, UGA or UAG) appropriately positioned at the end of the mRNA to be translated, with UAA and UAG preferred for mammalian or eukaryotic cell expression.
  • Expression vectors will preferably but optionally include at least one selectable marker.
  • markers include, e.g., but not limited to, methotrexate (MTX), dihydrofolate reductase (DHFR, US Pat.Nos. 4,399,216; 4,634,665; 4,656,134; 4,956,288; 5,149,636; 5,179,017, ampicillin, neomycin (G418), mycophenolic acid, or glutamine synthetase (GS, US Pat.Nos. 5,122,464; 5,770,359; 5,827,739) resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culturing in E.
  • MTX methotrexate
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • coli and other bacteria or prokaryotics (the above patents are entirely incorporated hereby by reference).
  • Appropriate culture mediums and conditions for the above- described host cells are known in the art. Suitable vectors will be readily apparent to the skilled artisan. Introduction of a vector construct into a host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid- mediated transfection, electroporation, transduction, infection or other known methods. Such methods are described in the art, such as Sambrook, supra, Chapters 1-4 and 16- 18; Ausubel, supra, Chapters 1, 9, 13, 15, 16.
  • At least one GLP-I mimetibody or specified portion or variant of the present invention can be expressed in a modified form, such as a fusion protein, and can include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, can be added to the N-terminus of a GLP-I mimetibody or specified portion or variant to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties can be added to a GLP-I mimetibody or specified portion or variant of the present invention to facilitate purification. Such regions can be removed prior to final preparation of a GLP-I mimetibody or at least one fragment thereof. Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 17.29-17.42 and 18.1- 18.74; Ausubel, supra, Chapters 16, 17 and 18.
  • Those of ordinary skill in the art are knowledgeable in the numerous expression systems available for expression of a nucleic acid encoding a protein of the present invention.
  • Illustrative of cell cultures useful for the production of the mimetibodies, specified portions or variants thereof, are mammalian cells. Mammalian cell systems often will be in the form of monolayers of cells although mammalian cell suspensions or bioreactors can also be used.
  • COS-I e.g., ATCC CRL 1650
  • COS-7 e.g., ATCC CRL-1651
  • HEK293, BHK21 e.g., ATCC CRL-10
  • CHO e.g., ATCC CRL 1610, DG-4
  • BSC-I e.g., ATCC CRL-26 cell lines
  • hepG2 cells P3X63Ag8.653, SP2/0-Agl4, 293 cells, HeLa cells and the like, which are readily available from, for example, American Type Culture Collection, Manassas, Va.
  • Preferred host cells include cells of lymphoid origin such as myeloma and lymphoma cells. Particularly preferred host cells are P3X63Ag8.653 cells (ATCC Accession Number CRL-1580) and SP2/0-Agl4 cells (ATCC Accession Number CRL- 1851).
  • Expression vectors for these cells can include one or more of the following expression control sequences, such as, but not limited to an origin of replication; a promoter (e.g., late or early SV40 promoters, the CMV promoter (e.g., US Pat.Nos. 5,168,062; 5,385,839), an HSV tk promoter, a pgk (phosphoglycerate kinase) promoter, an EF-I alpha promoter (e.g, US Pat.No.
  • a promoter e.g., late or early SV40 promoters, the CMV promoter (e.g., US Pat.Nos. 5,168,062; 5,385,839)
  • HSV tk promoter e.g., SV tk promoter
  • pgk phosphoglycerate kinase
  • EF-I alpha promoter e.g, US Pat.No.
  • At least one human immunoglobulin promoter at least one human immunoglobulin promoter; an enhancer, and/or processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site), and transcriptional terminator sequences.
  • an enhancer, and/or processing information sites such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site), and transcriptional terminator sequences.
  • polyadenlyation or transcription terminator sequences are typically incorporated into the vector.
  • An example of a terminator sequence is the polyadenlyation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript can also be included.
  • An example of a splicing sequence is the VPl intron from SV40 (Sprague, et al., J. Virol. 45:773-781 (1983)).
  • gene sequences to control replication in the host cell can be incorporated into the vector, as known in the art.
  • a GLP-I mimetibody or specified portion or variant can be recovered and purified from recombinant cell cultures by well-known methods including, but not limited to, protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography (“HPLC”) can also be employed for purification.
  • HPLC high performance liquid chromatography
  • Mimetibodies or specified portions or variants of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells.
  • a eukaryotic host including, for example, yeast, higher plant, insect and mammalian cells.
  • the GLP-I mimetibody or specified portion or variant of the present invention can be glycosylated or can be non-glycosylated, with glycosylated preferred.
  • Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Sections 17.37-17.42; Ausubel, supra, Chapters 10, 12, 13, 16, 18 and 20, Colligan, Protein Science, supra, Chapters 12-14, all entirely incorporated herein by reference.
  • the isolated mimetibodies of the present invention comprise a GLP-I mimetibody or specified portion or variant encoded by any one of the polynucleotides of the present invention as discussed more fully herein, or any isolated or prepared GLP-I mimetibody or specified portion or variant thereof.
  • the GLP-I mimetibody or ligand-binding portion or variant binds at least one GLP-I protein ligand and thereby provides at least one GLP-I biological activity of the corresponding protein or a fragment thereof.
  • Different therapeutically or diagnostically significant proteins are well known in the art and suitable assays or biological activities of such proteins are also well known in the art.
  • Non-limiting examples of suitable GLP-I peptides, variants and derivatives for this invention appear as SEQ ID NO:1: His-Xaa2-Xaa3-Gly-Xaa5-Xaa6-Xaa7-Xaa8- Xaa9-XaalO-Xaal 1-Xaal2-Xaal3-Xaal4-Xaal5-Xaal6-Xaal7-Xaal8-Xaal9-Xaa2 ⁇ -
  • Xaa21-Phe-Xaa23-Xaa24-Xaa25-Xaa26-Xaa27-Xaa28-Xaa29-Xaa30-Xaa31 wherein: Xaa2 is Ala, GIy, Ser, Thr, Leu, He, VaI, GIu, Asp, or Lys; Xaa3 is GIu, Asp, or Lys; Xaa5 is Thr, Ala, GIy, Ser, Leu, lie, VaI, Arg, His, GIu, Asp, or Lys; Xaa6 is Phe, His, Trp, or Tyr; Xaa7 is Thr or Asn; Xaa8 is Ser, Ala, GIy, Thr, Leu, lie, VaI, GIu, Asp, or Lys; Xaa9 is Asp or GIu; XaalO is VaI, Ala, GIy, Ser, Thr, Leu
  • GLP-I peptides, variants or derivatives are exemplied in SEQ ID NO:6: His-Xaa2-Xaa3-Gly-Thr-Xaa6- Xaa7-Xaa8-Xaa9-XaalO- Ser-Xaal2-Tyr-Xaal4-Glu-Xaal6-Xaal7-Xaal8-Xaal9-Lys-Xaa21-Phe-Xaa23-Ala- Trp-Leu-Xaa27-Xaa28-Gly-Xaa30, wherein: Xaa2 is Ala, GIy, or Ser; Xaa3 is GIu or Asp; Xaa6 is Phe or Tyr; Xaa7 is Thr or Asn; Xaa8 is Ser, Thr or Ala; Xaa9 is Asp or
  • GIu GIu
  • XaalO VaI, Leu, Met or He
  • Xaal2 Ser or Lys
  • Xaal4 is Leu, Ala or Met
  • Xaal ⁇ is GIy, Ala, GIu or Asp
  • Xaal7 is GIn or GIu
  • Xaal8 is Ala or Lys
  • Xaal9 is Ala, VaI, He, Leu or Met
  • Xaa21 is GIu or Leu
  • Xaa23 is He, Ala, VaI, Leu or GIu
  • Xaa27 is VaI or Lys
  • Xaa28 is Lys or Asn
  • Xaa30 is Arg or GIu.
  • DPP-IV dipeptidyl-peptidase IV
  • a GLP-I mimetibody, or specified portion or variant thereof, that partially or preferably substantially provides at least one GLP-I biological activity, can bind the GLP-I ligand and thereby provide at least one activity that is otherwise mediated through the binding of GLP-I to at least one ligand, such as a GLP-I receptor, or through other protein-dependent or mediated mechanisms.
  • GLP-I mimetibody activity refers to a GLP-I mimetibody that can modulate or cause at least one GLP-I dependent activity by about 20-10,000%, preferably by at least about 60, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 550, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 % or more, depending on the assay.
  • a human GLP- 1 mimetibody or specified portion or variant of the invention can be similar to any class (IgG, IgA, IgM, etc.) or isotype and can comprise at least a portion of a kappa or lambda light chain.
  • the human GLP-I mimetibody or specified portion or variant comprises IgG heavy chain variable fragments, hinge region, CH2 and CH3 of, at least one of isotypes, e.g., IgGl, IgG2, IgG3 or IgG4.
  • At least one GLP-I mimetibody or specified portion or variant of the invention binds at least one ligand, subunit, fragment, portion or any combination thereof.
  • the at least one GLP-I peptide, variant or derivative of at least one GLP-I mimetibody, specified portion or variant of the present invention can optionally bind at least one specified epitope of the ligand.
  • the binding epitope can comprise any combination of at least one amino acid sequence of at least 1-3 amino acids to the entire specified portion of contiguous amino acids of the sequences of a protein ligand, such as a GLP-I receptor or portion thereof.
  • Such mimetibodies can be prepared by j oining together the various portions of Formula (I) of the GLP-I mimetibody using known techniques, by preparing and expressing at least one nucleic acid molecules that encode the GLP-I mimetibody, using known techniques of recombinant DNA technology or by using any other suitable method, such as chemical synthesis.
  • Mimetibodies that bind to human GLP-I ligands, such as receptors, and that comprise a defined heavy or light chain variable region or portion thereof can be prepared using suitable methods, such as phage display (Katsube, Y., et al, Int JMol. Med, 1(5): 863-868 (1998)) or methods that employ transgenic animals, as known in the art.
  • the GLP-I mimetibody, specified portion or variant can be expressed using the encoding nucleic acid or portion thereof in a suitable host cell.
  • the invention also relates to mimetibodies, ligand-binding fragments and immunoglobulin chains comprising amino acids in a sequence that is substantially the same as an amino acid sequence described herein.
  • mimetibodies or ligand-binding fragments thereof can bind human GLP-I ligands, such as receptors, with high affinity (e.g., K D less than or equal to about 10 "7 M).
  • Amino acid sequences that are substantially the same as the sequences described herein include sequences comprising conservative amino acid substitutions, as well as amino acid deletions and/or insertions.
  • a conservative amino acid substitution refers to the replacement of a first amino acid by a second amino acid that has chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity/ hydrophilicity) that are similar to those of the first amino acid.
  • Conservative substitutions include replacement of one amino acid by another within the following groups: lysine (K), arginine (R) and histidine (H); aspartate (D) and glutamate (E); asparagine (N), glutamine (Q), serine (S), threonine (T), tyrosine (Y), K, R, H, D and E; alanine (A), valine (V), leucine (L), isoleucine (I), proline (P), phenylalanine (F), tryptophan (W), methionine (M), cysteine (C) and glycine (G); F, W and Y; C, S and T.
  • Amino Acid Codes are examples of amino acids.
  • amino acids that make up mimetibodies or specified portions or variants of the present invention are often abbreviated.
  • the amino acid designations can be indicated by designating the amino acid by its single letter code, its three letter code, name, or three nucleotide codon(s) as is well understood in the art (see Alberts, B., et al., Molecular Biology of The Cell, Third Ed., Garland Publishing, Inc., New York, 1994).
  • a GLP-I mimetibody or specified portion or variant of the present invention can include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation, as specified herein.
  • Such or other sequences that can be used in the present invention include, but are not limited to the following sequences presented in Table 1, as shown corresponding to specified portions of SEQ ID NOS:47-64, where the partial variable region of the antibody sequence can be, but is not limited to, at least one portion of at least one of SEQ ID NOS:47-55, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as further described in Figures 1-9 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ JD NOS: 1-9.
  • the CH2, CH3 and hinge region can be, but not limited to, at least one portion of at least one of SEQ ID NOS:56-64, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as as further described in Figures 32-40 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ JD NOS:32-40.
  • PCT publication WO 05/05604 PCT US04/19898 filed June 24, 2004 and published January 20, 2005, with corresponding SEQ JD NOS:32-40.
  • the number of amino acid substitutions a skilled artisan would make depends on many factors, including those described above.
  • the number of amino acid substitutions, insertions or deletions for at least one of a GLP-I mimetibody will not be more than 40, 30, 20,19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 amino acids, such as 1-30 or any range or value therein, as specified herein.
  • V, H, CH2, CH3 portions according to Formula I can be any suitable human or human compatable sequence, e.g., as presented in Table 1, where the partial variable region of the antibody sequence can be, but is not limited to, at least one portion of at least one of SEQ JD NOS:47-55, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion as further described in Figures 1-9 of PCT publication WO 05/05604 (PCT US04/19898) filed June 24, 2004 and published January 20, 2005, with corresponding SEQ JD NOS: 1-9; and where the CH2, CH3 and hinge region can be, but not limited to, at least one portion of at least one of SEQ JD NOS: 56-64, or fragment thereof as described in Table 1, further optionally comprising at least one substitution, insertion or deletion
  • the P portion can comprise at least one GLP-I therapeutic peptide known in the art or described herein, such as, but not limited to those presented in SEQ ID NO: 1, or any combination or consensus sequence thereof, or any fusion protein thereof.
  • the P portion can comprise at least one GLP-I peptide having the the sequence of at least one of SEQ ID NO:6, or any combination or consensus sequence thereof, or any fusion protein thereof.
  • the optional linker sequence can be any suitable peptide linker as known in the art. Preferred sequences include any combination of G and S, e.g., X1-X2-X3-X4-...- Xn, where X can be G or S, and n can be 5-30.
  • Non-limiting examples include GS, GGS, GGGS (SEQ ID NO: 16), GSGGGS (SEQ ID NO: 17), GGSGGGS (SEQ ID NO: 18), GGSGGGSGG (SEQ ID NO: 19) and GGGSGGGSGG (SEQ ID NO:20); and the like.
  • Amino acids in a GLP-I mimetibody or specified portion or variant of the present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, Science 244:1081-1085 (1989)).
  • site-directed mutagenesis or alanine-scanning mutagenesis e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, Science 244:1081-1085 (1989)
  • the latter procedure introduces single alanine mutations at every residue in the molecule.
  • the resulting mutant molecules are then tested for biological activity, such as, but not limited to at least one protein related activity, as specified herein or as known in the art.
  • Sites that are critical for GLP-I mimetibody or specified portion or variant binding can also be identified by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith, et al., J. MoI. Biol. 224:899-904
  • Mimetibodies or specified portions or variants of the present invention can comprise as the P portion of Formula (I), e.g. but not limited to, at least one portion of at least one of SEQ ID NOS: 1 and 6.
  • a GLP-I mimetibody or specified portion or variant can further optionally comprise at least one functional portion of at least one polypeptide as P portion of Formula (I), at least 90-100% of at least on of SEQ ID NOS: 1 and 6.
  • Non-limiting variants that can enhance or maintain at least one of the listed activities above include, but are not limited to, any of the above polypeptides, further comprising at least one mutation corresponding to at least one substitution, insertion or deletion that does not significantly affect the suitable biological activtities or functions of said GLP-I mimetibody.
  • the P amino acid sequence, or portion thereof has about 90-100% identity (i.e., 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any range or value therein) to the corresponding amino acid sequence of the corresponding portion of at least one of SEQ ID NOS : 1 and 6.
  • 90-100% amino acid identity i.e., 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any range or value therein
  • 90-100% amino acid identity is determined using a suitable computer algorithm, as known in the art.
  • Mimetibodies or specified portions or variants of the present invention can comprise any number of contiguous amino acid residues from a GLP-I mimetibody or specified portion or variant of the present invention, wherein that number is selected from the group of integers consisting of from 10-100% of the number of contiguous residues in a GLP-I mimetibody.
  • this subsequence of contiguous amino acids is at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250 or more amino acids in length, or any range or value therein.
  • the number of such subsequences can be any integer selected from the group consisting of from 1 to 20, such as at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more.
  • the present invention includes at least one biologically active GLP-I mimetibody or specified portion or variant of the present invention.
  • Biologically active mimetibodies or specified portions or variants have a specific activity at least 20%, 30%, or 40%, and preferably at least 50%, 60%, or 70%, and most preferably at least 80%, 90%, or 95%-1000% of that of the native (non- synthetic), endogenous or related and known inserted or fused protein or specified portion or variant.
  • Methods of assaying and quantifying measures of enzymatic activity and substrate specificity are well known to those of skill in the art.
  • the invention relates to human mimetibodies and ligand- binding fragments, as described herein, which are modified by the covalent attachment of an organic moiety.
  • modification can produce a GLP-I mimetibody or ligand- binding fragment with improved pharmacokinetic properties (e.g., increased in vivo serum half-life).
  • the organic moiety can be a linear or branched hydrophilic polymeric group, fatty acid group, or fatty acid ester group.
  • the hydrophilic polymeric group can have a molecular weight of about 800 to about 120,000 Daltons and can be a polyalkane glycol (e.g., polyethylene glycol (PEG), polypropylene glycol (PPG)), carbohydrate polymer, amino acid polymer or polyvinyl pyrolidone, and the fatty acid or fatty acid ester group can comprise from about eight to about forty carbon atoms.
  • a polyalkane glycol e.g., polyethylene glycol (PEG), polypropylene glycol (PPG)
  • carbohydrate polymer e.g., amino acid polymer or polyvinyl pyrolidone
  • the fatty acid or fatty acid ester group can comprise from about eight to about forty carbon atoms.
  • the modified mimetibodies and ligand-binding fragments of the invention can comprise one or more organic moieties that are covalently bonded, directly or indirectly, to the GLP-I mimetibody or specified portion or variant.
  • Each organic moiety that is bonded to a GLP-I mimetibody or ligand-binding fragment of the invention can independently be a hydrophilic polymeric group, a fatty acid group or a fatty acid ester group.
  • fatty acid encompasses mono-carboxylic acids and di-carboxylic acids.
  • Hydrophilic polymers suitable for modifying mimetibodies of the invention can be linear or branched and include, for example, polyalkane glycols (e.g., PEG, monomethoxy- polyethylene glycol (mPEG), PPG and the like), carbohydrates (e.g., dextran, cellulose, oligosaccharides, polysaccharides and the like), polymers of hydrophilic amino acids (e.g., polylysine, polyarginine, polyaspartate and the like), polyalkane oxides (e.g., polyethylene oxide, polypropylene oxide and the like) and polyvinyl pyrolidone.
  • polyalkane glycols e.g., PEG, monomethoxy- polyethylene glycol (mPEG), PPG and the like
  • carbohydrates e.g., dextran, cellulose, oligosaccharides, polysaccharides and the like
  • polymers of hydrophilic amino acids e.g., polylys
  • the hydrophilic polymer that modifies the GLP-I mimetibody of the invention has a molecular weight of about 800 to about 150,000 Daltons as a separate molecular entity.
  • PEG 250 O, PEG 50 oo, PEG 75 Oo, PEGgooo, PEG 100 Oo, PEG 12500 , PEG 150OO , and PEG 2 o,ooo, wherein the subscript is the average molecular weight of the polymer in Daltons can be used.
  • the hydrophilic polymeric group can be substituted with one to about six alkyl, fatty acid or fatty acid ester groups.
  • Hydrophilic polymers that are substituted with a fatty acid or fatty acid ester group can be prepared by employing suitable methods. For example, a polymer comprising an amine group can be coupled to a carboxylate of the fatty acid or fatty acid ester, and an activated carboxylate (e.g., activated with N 5 N- carbonyl diimidazole) on a fatty acid or fatty acid ester can be coupled to a hydroxyl group on a polymer.
  • an activated carboxylate e.g., activated with N 5 N- carbonyl diimidazole
  • Fatty acids and fatty acid esters suitable for modifying mimetibodies of the invention can be saturated or can contain one or more units of unsaturation.
  • Fatty acids that are suitable for modifying mimetibodies of the invention include, for example, n- dodecanoate (C 12 , laurate), n-tetradecanoate (C 14 , myristate), n-octadecanoate (C 18 , stearate), n-eicosanoate (C 2 o, arachidate), n-docosanoate (C 22 , behenate), n- triacontanoate (C 30 ), n-tetracontanoate (C 40 ), cw- ⁇ 9-octadecanoate (C 18 , oleate), all cis- ⁇ 5,8,l l,14-eicosatetraenoate (C 20 , arachidonate), octanedioic acid,
  • the modified human mimetibodies and ligand-binding fragments can be prepared using suitable methods, such as by reaction with one or more modifying agents.
  • An "activating group” is a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group.
  • amine-reactive activating groups include electrophilic groups such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl esters (NHS), and the like.
  • Activating groups that can react with thiols include, for example, maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB-thiol), and the like.
  • An aldehyde functional group can be coupled to amine- or hydrazide- containing molecules, and an azide group can react with a trivalent phosphorous group to form phosphoramidate or phosphorimide linkages.
  • Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., Bioconjugate Techniques, Academic Press: San Diego, CA (1996)).
  • An activating group can be bonded directly to the organic group (e.g., hydrophilic polymer, fatty acid, fatty acid ester), or through a linker moiety, for example a divalent C 1 -C 12 group wherein one or more carbon atoms can be replaced by a heteroatom such as oxygen, nitrogen or sulfur.
  • Suitable linker moieties include, for example, tetraethylene glycol, - (CHiO 3 -, -NH-(CH 2 ) 6 -NH-, -(CH 2 ) 2 -NH- and -CH 2 -O-CH 2 -CH 2 -O-CH 2 -CH 2 -O-CH-NH- .
  • Modifying agents that comprise a linker moiety can be produced, for example, by reacting a mono-Boc-alkyldiamine (e.g., mono-Boc-ethylenediamine, mono-Boc- diaminohexane) with a fatty acid in the presence of l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) to form an amide bond between the free amine and the fatty acid carboxylate.
  • a mono-Boc-alkyldiamine e.g., mono-Boc-ethylenediamine, mono-Boc- diaminohexane
  • EDC l-ethyl-3-(3-dimethylaminopropyl) carbodiimide
  • the Boc protecting group can be removed from the product by treatment with trifluoroacetic acid (TFA) to expose a primary amine that can be coupled to another carboxylate as described, or can be reacted with maleic anhydride and the resulting product cyclized to produce an activated maleimido derivative of the fatty acid.
  • TFA trifluoroacetic acid
  • the modified mimetibodies of the invention can be produced by reacting an human GLP-I mimetibody or ligand-binding fragment with a modifying agent.
  • the organic moieties can be bonded to the GLP-I mimetibody in a non-site specific manner by employing an amine-reactive modifying agent, for example, an NHS ester of PEG.
  • Modified human mimetibodies or ligand-binding fragments can also be prepared by reducing disulfide bonds (e.g., intra-chain disulfide bonds) of a GLP-I mimetibody or ligand-binding fragment.
  • the reduced GLP-I mimetibody or ligand- binding fragment can then be reacted with a thiol-reactive modifying agent to produce the modified GLP-I mimetibody of the invention.
  • Modified human mimetibodies and ligand-binding fragments comprising an organic moiety that is bonded to specific sites of a GLP-I mimetibody or specified portion or variant of the present invention can be prepared using suitable methods, such as reverse proteolysis (Fisch et ah, Bioconjugate Chem., 3:147-153 (1992); Werlen et al, Bioconjugate Chem., 5:411-417 (1994); Kumaran et al, Protein ScL 6(10): 2233 -2241 (1997); Itoh et al, Bioorg.
  • the present invention also provides at least one GLP-I mimetibody or specified portion or variant composition comprising at least one, at least two, at least three, at least four, at least five, at least six or more mimetibodies or specified portions or variants thereof, as described herein and/or as known in the art that are provided in a non-naturally occurring composition, mixture or form.
  • Such composition percentages are by weight, volume, concentration, molarity, or molality as liquid or dry solutions, mixtures, suspension, emulsions or colloids, as known in the art or as described herein.
  • compositions can comprise 0.00001-99.9999 percent by weight, volume, concentration, molarity, or molality as liquid, gas, or dry solutions, mixtures, suspension, emulsions or colloids, as known in the art or as described herein, on any range or value therein, such as but not limited to 0.00001, 0.00003, 0.00005, 0.00009,
  • the composition can optionally further comprise an effective amount of at least one compound or protein selected from at least one of a diabetes or insuling metabolism related drug, an anti-infective drug, a cardiovascular (CV) system drug, a central nervous system (CNS) drug, an autonomic nervous system (ANS) drug, a respiratory tract drug, a gastrointestinal (GI) tract drug, a hormonal drug, a drug for fluid or electrolyte balance, a hematologic drug, an antineoplactic, an immunomodulation drug, an ophthalmic, otic or nasal drug, a topical drug, a nutritional drug or the like.
  • CV cardiovascular
  • CNS central nervous system
  • ANS autonomic nervous system
  • a respiratory tract drug a gastrointestinal (GI) tract drug
  • a hormonal drug a drug for fluid or electrolyte balance
  • a hematologic drug an antineoplactic
  • an immunomodulation drug an ophthalmic, otic or nasal drug
  • topical drug a nutritional drug or the like.
  • Such drugs are well known in the art, including formulations, indications, dosing and administration for each presented herein (see e.g., Nursing 2001 Handbook of Drugs, 21 st edition, Springhouse Corp., Springhouse, PA, 2001; Health Professional's Drug Guide 2001, ed., Shannon, Wilson, Stang, Prentice-Hall, Inc, Upper Saddle River, NJ; Pharmcotherapy Handbook, Wells et al., ed., Appleton & Lange, Stamford, CT, each entirely incorporated herein by reference).
  • the diabetes related drug can be at least one of glitazones, insulin and derivatives, sulfonylureas, meglitinides, biguanides, alpha-glucosidase inhibitors, protein tyrosine phosphastase-lB, glycogen synthase kinase 3, gluconeogenesis inhibitors, pyruvate dehydrogenase kinase (PDH) inhibitors, lipolysis inhibitors, fat oxidation inhibitors, carnitine palmitoyltransferase I and/or II inhibitors, beta-3 adrenoceptor agonists, sodium and glucose cotransporter (SGLT) inhibitors, or compounds that act on one or more of at least one of: autoimmune suppression, immune regulation, activation, proliferation, migration and/or suppressor cell function of T-cells, inhibition of T cell receptor/peptide/MHC-H interaction, Induction of T cell anergy, deletion of autoreactive T cells, reduction
  • the anti-infective drug can be at least one selected from amebicides or antiprotozoals, anthelmintics, antifungals, antimalarials, antituberculotics or antileprotics, aminoglycosides, penicillins, cephalosporins, tetracyclines, sulfonamides, fluoroquinolones, antivirals, macrolide anti-infectives and miscellaneous anti-infectives.
  • the CV drug can be at least one selected from inotropics, antiarrhythmics, antianginals, antihypertensives, antilipemics and miscellaneous cardiovascular drugs.
  • the CNS drug can be at least one selected from nonnarcotic analgesics or at least one selected from antipyretics, nonsteroidal anti-inflammatory drugs, narcotic or opiod analgesics, sedative-hypnotics, anticonvulsants, antidepressants, antianxiety drugs, antipsychotics, central nervous system stimulants, antiparkinsonians and miscellaneous central nervous system drugs.
  • the ANS drug can be at least one selected from cholinergics (parasympathomimetics), anticholinergics, adrenergics (sympathomimetics), adrenergic blockers (sympatholytics), skeletal muscle relaxants and neuromuscular blockers.
  • the respiratory tract drug can be at least one selected from antihistamines, bronchodilators, expectorants or antitussives and miscellaneous respiratory drugs.
  • the GI tract drug can be at least one selected from antacids, adsorbents, antiflatulents, digestive enzymes, gallstone solubilizers, antidiarrheals, laxatives, antiemetics and antiulcer drugs.
  • the hormonal drug can be at least one selected from corticosteroids, androgens, anabolic steroids, estrogens, progestins, gonadotropins, antidiabetic drugs, at least one glucagon, thyroid hormones, thyroid hormone antagonists, pituitary hormones and parathyroid-like drugs.
  • the drug for fluid and electrolyte balance can be at least one selected from diuretics, electrolytes, replacement solutions, acidif ⁇ ers and alkalinizers.
  • the hematologic drug can be at least one selected from hematinics, anticoagulants, blood derivatives and thrombolytic enzymes.
  • the antineoplastics can be at least one selected from alkylating drugs, antimetabolites, antibiotic antineoplastics, antineoplastics that alter hormone balance and miscellaneous antineoplastics.
  • the immunomodulation drug can be at least one selected from immunosuppressants, vaccines, toxoids, antitoxins, antivenins, immune serums and biological response modifiers.
  • the ophthalmic, otic, and nasal drugs can be at least one selected from ophthalmic anti-infectives, ophthalmic antiinflammatories, miotics, mydriatics, ophthalmic vasoconstrictors and miscellaneous ophthalmics, otics, nasal drugs.
  • the topical drug can be at least one selected from local anti-infectives, scabicides, pediculicides and topical corticosteroids.
  • the nutritional drug can be at least one selected from vitamins, minerals and calorics. See, e.g., contents of Nursing 2001 Drug Handbook, supra.
  • the at least one amebicide or antiprotozoal can be at least one selected from atovaquone, chloroquine hydrochloride, chloroquine phosphate, metronidazole, metronidazole hydrochloride and pentamidine isethionate.
  • the at least one anthelmintic can be at least one selected from mebendazole, pyrantel pamoate and thiabendazole.
  • the at least one antifungal can be at least one selected from amphotericin B, amphotericin B cholesteryl sulfate complex, amphotericin B lipid complex, amphotericin B liposomal, fluconazole, flucytosine, griseofulvin microsize, griseofulvin ultramicrosize, itraconazole, ketoconazole, nystatin and terbinafine hydrochloride.
  • the at least one antimalarial can be at least one selected from chloroquine hydrochloride, chloroquine phosphate, doxycycline, hydroxychloroquine sulfate, mefloquine hydrochloride, primaquine phosphate, pyrimethamine and pyrimethamine with sulfadoxine.
  • the at least one antituberculotic or antileprotic can be at least one selected from clofazimine, cycloserine, dapsone, ethambutol hydrochloride, isoniazid, pyrazinamide, rifabutin, rifampin, rifapentine and streptomycin sulfate.
  • the at least one aminoglycoside can be at least one selected from amikacin sulfate, gentamicin sulfate, neomycin sulfate, streptomycin sulfate and tobramycin sulfate.
  • the at least one penicillin can be at least one selected from amoxcillin/clavulanate potassium, amoxicillin trihydrate, ampicillin, ampicillin sodium, ampicillin trihydrate, ampicillin sodium/sulbactam sodium, cloxacillin sodium, dicloxacillin sodium, mezlocillin sodium, nafcillin sodium, oxacillin sodium, penicillin G benzathine, penicillin G potassium, penicillin G procaine, penicillin G sodium, penicillin V potassium, piperacillin sodium, piperacillin sodium/tazobactam sodium, ticarcillin disodium and ticarcillin disodium/clavulanate potassium.
  • the at least one cephalosporin can be at least one selected from at least one of cefaclor, cefadroxil, cefazolin sodium, cefdinir, cefepime hydrochloride, cefixime, cefmetazole sodium, cefonicid sodium, cefoperazone sodium, cefotaxime sodium, cefotetan disodium, cefoxitin sodium, cefpodoxime proxetil, cefprozil, ceftazidime, ceftibuten, ceftizoxime sodium, ceftriaxone sodium, cefuroxime axetil, cefuroxime sodium, cephalexin hydrochloride, cephalexin monohydrate, cephradine, loracarbef.
  • the at least one tetracycline can be at least one selected from demeclocycline hydrochloride, doxycycline calcium, doxycycline hyclate, doxycycline hydrochloride, doxycycline monohydrate, minocycline hydrochloride, tetracycline hydrochloride.
  • the at least one sulfonamide can be at least one selected from co-trimoxazole, sulfadiazine, sulfamethoxazole, sulfisoxazole, sulfisoxazole acetyl.
  • the at least one fluoroquinolone can be at least one selected from alatrofloxacin mesylate, ciprofloxacin, enoxacin, levofloxacin, lomefloxacin hydrochloride, nalidixic acid, norfloxacin, ofloxacin, sparfloxacin, trovafloxacin mesylate.
  • the at least one fluoroquinolone can be at least one selected from alatrofloxacin mesylate, ciprofloxacin, enoxacin, levofloxacin, lomefloxacin hydrochloride, nalidixic acid, norfloxacin, ofloxacin, sparfloxacin, trovafloxacin mesylate.
  • the at least one antiviral can be at least one selected from abacavir sulfate, acyclovir sodium, amantadine hydrochloride, amprenavir, cidofovir, delavirdine mesylate, didanosine, efavirenz, famciclovir, fomivirsen sodium, foscarnet sodium, ganciclovir, indinavir sulfate, lamivudine, lamivudine/zidovudine, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, rimantadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valacyclovir hydrochloride, zalcitabine, zanamivir, zidovudine.
  • the at least one macroline anti-infective can be at least one selected from azithromycin, clarithromycin, dirithromycin, erythromycin base, erythromycin estolate, erythromycin ethylsuccinate, erythromycin lactobionate, erythromycin stearate.
  • the at least one miscellaneous anti-infective can be at least one selected from aztreonam, bacitracin, chloramphenicol sodium sucinate, clindamycin hydrochloride, clindamycin palmitate hydrochloride, clindamycin phosphate, imipenem and cilastatin sodium, meropenem, nitrofurantoin macrocrystals, nitrofurantoin microcrystals, quinupristin/dalfopristin, spectinomycin hydrochloride, trimethoprim, vancomycin hydrochloride. (See, e.g., pp.
  • the at least one inotropic can be at least one selected from amrinone lactate, digoxin, milrinone lactate.
  • the at least one antiarrhythmic can be at least one selected from adenosine, amiodarone hydrochloride, atropine sulfate, bretylium tosylate, diltiazem hydrochloride, disopyramide, disopyramide phosphate, esmolol hydrochloride, flecainide acetate, ibutilide fumarate, lidocaine hydrochloride, mexiletine hydrochloride, moricizine hydrochloride, phenytoin, phenytoin sodium, procainamide hydrochloride, propafenone hydrochloride, propranolol hydrochloride, quinidine bisulfate, quinidine gluconate, quinidine polygalacturonate, quinidine sulfate,
  • the at least one antianginal can be at least one selected from amlodipidine besylate, amyl nitrite, bepridil hydrochloride, diltiazem hydrochloride, isosorbide dinitrate, isosorbide mononitrate, nadolol, nicardipine hydrochloride, nifedipine, nitroglycerin, propranolol hydrochloride, verapamil, verapamil hydrochloride.
  • the at least one antihypertensive can be at least one selected from acebutolol hydrochloride, amlodipine besylate, atenolol, benazepril hydrochloride, betaxolol hydrochloride, bisoprolol fumarate, candesartan cilexetil, captopril, carteolol hydrochloride, carvedilol, clonidine, clonidine hydrochloride, diazoxide, diltiazem hydrochloride, doxazosin mesylate, enalaprilat, enalapril maleate, eprosartan mesylate, felodipine, fenoldopam mesylate, fosinopril sodium, guanabenz acetate, guanadrel sulfate, guanfacine hydrochloride, hydralazine hydrochloride, irbe
  • the at least one miscellaneous CV drug can be at least one selected from abciximab, alprostadil, arbutamine hydrochloride, cilostazol, clopidogrel bisulfate, dipyridamole, eptifibatide, midodrine hydrochloride, pentoxifylline, ticlopidine hydrochloride, tirofiban hydrochloride. (See, e.g., pp. 215-336 of Nursing
  • the at least one nonnarcotic analgesic or antipyretic can be at least one selected from acetaminophen, aspirin, choline magnesium trisalicylate, diflunisal, magnesium salicylate.
  • the at least one nonsteroidal anti-inflammatory drug can be at least one selected from celecoxib, diclofenac potassium, diclofenac sodium, etodolac, fenoprofen calcium, flurbiprofen, ibuprofen, indomethacin, indomethacin sodium trihydrate, ketoprofen, ketorolac tromethamine, nabumetone, naproxen, naproxen sodium, oxaprozin, pirpxicam, rofecoxib, sulindac.
  • the at least one narcotic or opiod analgesic can be at least one selected from alfentanil hydrochloride, buprenorphine hydrochloride, butorphanol tartrate, codeine phosphate, codeine sulfate, fentanyl citrate, fentanyl transdermal system, fentanyl transmucosal, hydromorphone hydrochloride, meperidine hydrochloride, methadone hydrochloride, morphine hydrochloride, morphine sulfate, morphine tartrate, nalbuphine hydrochloride, oxycodone hydrochloride, oxycodone pectinate, oxymorphone hydrochloride, pentazocine hydrochloride, pentazocine hydrochloride and naloxone hydrochloride, pentazocine lactate, propoxyphene hydrochloride, propoxyphene napsylate, remifentanil hydrochloride, sufentanil
  • the at least one sedative-hypnotic can be at least one selected from chloral hydrate, estazolam, flurazepam hydrochloride, pentobarbital, pentobarbital sodium, phenobarbital sodium, secobarbital sodium, temazepam, triazolam, zaleplon, Zolpidem tartrate.
  • the at least one anticonvulsant can be at least one selected from acetazolamide sodium, carbamazepine, clonazepam, clorazepate dipotassium, diazepam, divalproex sodium, ethosuximde, fosphenytoin sodium, gabapentin, lamotrigine, magnesium sulfate, phenobarbital, phenobarbital sodium, phenytoin, phenytoin sodium, phenytoin sodium (extended), primidone, tiagabine hydrochloride, topiramate, valproate sodium, valproic acid.
  • the at least one antidepressant can be at least one selected from amitriptyline hydrochloride, amitriptyline pamoate, amoxapine, bupropion hydrochloride, citalopram hydrobromide, clomipramine hydrochloride, desipramine hydrochloride, doxepin hydrochloride, fluoxetine hydrochloride, imipramine hydrochloride, imipramine pamoate, mirtazapine, nefazodone hydrochloride, nortriptyline hydrochloride, paroxetine hydrochloride, phenelzine sulfate, sertraline hydrochloride, tranylcypromine sulfate, trimipramine maleate, venlafaxine hydrochloride.
  • the at least one antianxiety drug can be at least one selected from alprazolam, buspirone hydrochloride, chlordiazGLP-lxide, chlordiazGLP-lxide hydrochloride, clorazepate dipotassium, diazepam, doxepin hydrochloride, hydroxyzine embonate, hydroxyzine hydrochloride, hydroxyzine pamoate, lorazepam, mephrobamate, midazolam hydrochloride, oxazepam.
  • the at least one antipsychotic drug can be at least one selected from chlorpromazine hydrochloride, clozapine, fluphenazine decanoate, fluephenazine enanthate, fluphenazine hydrochloride, haloperidol, haloperidol decanoate, haloperidol lactate, loxapine hydrochloride, loxapine succinate, mesoridazine besylate, molindone hydrochloride, olanzapine, perphenazine, pimozide, prochlorperazine, quetiapine fumarate, risperidone, thioridazine hydrochloride, thiothixene, thiothixene hydrochloride, trifluoperazine hydrochloride.
  • the at least one central nervous system stimulant can be at least one selected from amphetamine sulfate, caffeine, dextroamphetamine sulfate, doxapram hydrochloride, methamphetamine hydrochloride, methylphenidate hydrochloride, modafinil, pemoline, phentermine hydrochloride.
  • the at least one antiparkinsonian can be at least one selected from amantadine hydrochloride, benztropine mesylate, biperiden hydrochloride, biperiden lactate, bromocriptine mesylate, carbidopa-levodopa, entacapone, levodopa, pergolide mesylate, pramipexole dihydrochloride, ropinirole hydrochloride, selegiline hydrochloride, tolcapone, trihexyphenidyl hydrochloride.
  • the at least one miscellaneous central nervous system drug can be at least one selected from bupropion hydrochloride, donepezil hydrochloride, droperidol, fluvoxamine maleate, lithium carbonate, lithium citrate, naratriptan hydrochloride, nicotine polacrilex, nicotine transdermal system, propofol, rizatriptan benzoate, sibutramine hydrochloride monohydrate, sumatriptan succinate, tacrine hydrochloride, zolmitriptan. (See, e.g., pp.
  • the at least one cholinergic can be at least one selected from bethanechol chloride, edrophonium chloride, neostigmine bromide, neostigmine methylsulfate, physostigmine salicylate, pyridostigmine bromide.
  • the at least one anticholinergics can be at least one selected from atropine sulfate, dicyclomine hydrochloride, glycopyrrolate, hyoscyamine, hyoscyamine sulfate, propantheline bromide, scopolamine, scopolamine butylbromide, scopolamine hydrobromide.
  • the at least one adrenergics can be at least one selected from dobutamine hydrochloride, dopamine hydrochloride, metaraminol bitartrate, norepinephrine bitartrate, phenylephrine hydrochloride, pseudoephedrine hydrochloride, pseudoephedrine sulfate.
  • the at least one adrenergic blocker can be at least one selected from dihydroergotamine mesylate, ergotamine tartrate, methysergide maleate, propranolol hydrochloride.
  • the at least one skeletal muscle relaxant can be at least one selected from baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine hydrochloride, dantrolene sodium, methocarbamol, tizanidine hydrochloride.
  • the at least one neuromuscular blockers can be at least one selected from atracurium besylate, cisatracurium besylate, doxacurium chloride, mivacurium chloride, pancuronium bromide, pipecuronium bromide, rapacuronium bromide, rocuronium bromide, succinylcholine chloride, tubocurarine chloride, vecuronium bromide. (See, e.g., pp. 531 -84 of Nursing 2001 Drug Handbook.)
  • the at least one antihistamine can be at least one selected from brompheniramine maleate, cetirizine hydrochloride, chlorpheniramine maleate, clemastine fumarate, cyproheptadine hydrochloride, diphenhydramine hydrochloride, fexofenadine hydrochloride, loratadine, promethazine hydrochloride, promethazine theoclate, triprolidine hydrochloride.
  • the at least one bronchodilators can be at least one selected from albuterol, albuterol sulfate, aminophylline, atropine sulfate, ephedrine sulfate, epinephrine, epinephrine bitartrate, epinephrine hydrochloride, ipratropium bromide, isoproterenol, isoproterenol hydrochloride, isoproterenol sulfate, levalbuterol hydrochloride, metaproterenol sulfate, oxtriphylline, pirbuterol acetate, salmeterol xinafoate, terbutaline sulfate, theophylline.
  • the at least one expectorants or antitussives can be at least one selected from benzonatate, codeine phosphate, codeine sulfate, dextramethorphan hydrobromide, diphenhydramine hydrochloride, guaifenesin, hydromorphone hydrochloride.
  • the at least one miscellaneous respiratory drug can be at least one selected from acetylcysteine, beclomethasone dipropionate, beractant, budesonide, calfactant, cromolyn sodium, dornase alfa, GLP-lprostenol sodium, flunisolide, fluticasone propionate, montelukast sodium, nedocromil sodium, palivizumab, triamcinolone acetonide, zafirlukast, zileuton. (See, e.g., pp. 585-642 of Nursing 2001 Drug Handbook.)
  • the at least one antacid, adsorbents, or antiflatulents can be at least one selected from aluminum carbonate, aluminum hydroxide, calcium carbonate, magaldrate, magnesium hydroxide, magnesium oxide, simethicone, and sodium bicarbonate.
  • the at least one digestive enymes or gallstone solubilizers can be at least one selected from pancreatin, pancrelipase, and ursodiol.
  • the at least one antidiarrheal can be at least one selected from attapulgite, bismuth subsalicylate, calcium polycarbophil, diphenoxylate hydrochloride or atropine sulfate, loperamide, octreotide acetate, opium tincture, opium tincure (camphorated).
  • the at least one laxative can be at least one selected from bisocodyl, calcium polycarbophil, cascara sagrada, cascara sagrada aromatic fluidextract, cascara sagrada fluidextract, castor oil, docusate calcium, docusate sodium, glycerin, lactulose, magnesium citrate, magnesium hydroxide, magnesium sulfate, methylcellulose, mineral oil, polyethylene glycol or electrolyte solution, psyllium, senna, sodium phosphates.
  • the at least one antiemetic can be at least one selected from chlorpromazine hydrochloride, dimenhydrinate, dolasetron mesylate, dronabinol, granisetron hydrochloride, meclizine hydrochloride, metocloproamide hydrochloride, ondansetron hydrochloride, perphenazine, prochlorperazine, prochlorperazine edisylate, prochlorperazine maleate, promethazine hydrochloride, scopolamine, thiethylperazine maleate, trimethobenzamide hydrochloride.
  • the at least one antiulcer drug can be at least one selected from cimetidine, cimetidine hydrochloride, famotidine, lansoprazole, misoprostol, nizatidine, omeprazole, rabeprozole sodium, rantidine bismuth citrate, ranitidine hydrochloride, sucralfate. (See, e.g., pp.
  • the at least one coricosteroids can be at least one selected from betamethasone, betamethasone acetate or betamethasone sodium phosphate, betamethasone sodium phosphate, cortisone acetate, dexamethasone, dexamethasone acetate, dexamethasone sodium phosphate, fludrocortisone acetate, hydrocortisone, hydrocortisone acetate, hydrocortisone cypionate, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, methylprednisolone, methylprednisolone acetate, methylprednisolone sodium succinate, prednisolone, prednisolone acetate, prednisolone sodium phosphate, prednisolone tebutate, prednisone, triamcinolone, triamcinolone acetonide, triamcinolone diacetate.
  • the at least one androgen or anabolic steroids can be at least one selected from danazol, fluoxymesterone, methyltestosterone, nandrolone decanoate, nandrolone phenpropionate, testosterone, testosterone cypionate, testosterone enanthate, testosterone propionate, testosterone transdermal system.
  • the at least one estrogen or progestin can be at least one selected from esterified estrogens, estradiol, estradiol cypionate, estradiol/norethindrone acetate transdermal system, estradiol valerate, estrogens (conjugated), estropipate, ethinyl estradiol, ethinyl estradiol and desogestrel, ethinyl estradiol and ethynodiol diacetate, ethinyl estradiol and desogestrel, ethinyl estradiol and ethynodiol diacetate, ethinyl estradiol and levonorgestrel, ethinyl estradiol and norethindrone, ethinyl estradiol and norethindrone acetate, ethinyl estradiol and norgestimate, ethiny
  • the at least one gonadroptropin can be at least one selected from ganirelix acetate, gonadoreline acetate, histrelin acetate, menotropins.
  • the at least one antidiabetic or glucaon can be at least one selected from acarbose, chlorpropamide, glimepiride, glipizide, glucagon, glyburide, insulins, metformin hydrochloride, miglitol, pioglitazone hydrochloride, repaglinide, rosiglitazone maleate, troglitazone.
  • the at least one thyroid hormone can be at least one selected from levothyroxine sodium, liothyronine sodium, liotrix, thyroid.
  • the at least one thyroid hormone antagonist can be at least one selected from methimazole, potassium iodide, potassium iodide (saturated solution), propylthiouracil, radioactive iodine (sodium iodide 131 I), strong iodine solution.
  • the at least one pituitary hormone can be at least one selected from corticotropin, cosyntropin, desmopressin acetate, leuprolide acetate, rGLP-lsitory corticotropin, somatrem, somatropin, vasopressin.
  • the at least one parathyroid-like drug can be at least one selected from calcifediol, calcitonin (human), calcitonin (salmon), calcitriol, dihydrotachysterol, etidronate disodium. (See, e.g., pp. 696-796 of Nursing 2001 Drug Handbook.)
  • the at least one diuretic can be at least one selected from acetazolamide, acetazolamide sodium, amiloride hydrochloride, bumetanide, chlorthalidone, ethacrynate sodium, ethacrynic acid, furosemide, hydrochlorothiazide, indapamide, mannitol, metolazone, spironolactone, torsemide, triamterene, urea.
  • the at least one electrolyte or replacement solution can be at least one selected from calcium acetate, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, calcium lactate, calcium phosphate (dibasic), calcium phosphate (tribasic), dextran (high-molecular-weight), dextran (low-molecular-weight), hetastarch, magnesium chloride, magnesium sulfate, potassium acetate, potassium bicarbonate, potassium chloride, potassium gluconate, Ringer's injection, Ringer's injection (lactated), sodium chloride.
  • the at least one acidifier or alkalinizer can be at least one selected from sodium bicarbonate, sodium lactate, tromethamine. (See, e.g., pp. 797-833 of Nursing 2001 Drug Handbook.)
  • the at least one hematinic can be at least one selected from ferrous fumarate, ferrous gluconate, ferrous sulfate, ferrous sulfate (dried), iron dextran, iron sorbitol, polysaccharide-iron complex, sodium ferric gluconate complex.
  • the at least one anticoagulant can be at least one selected from ardeparin sodium, dalteparin sodium, danaparoid sodium, enoxaparin sodium, heparin calcium, heparin sodium, warfarin sodium.
  • the at least one blood derivative can be at least one selected from albumin 5%, albumin 25%, antihemophilic factor, anti-inhibitor coagulant complex, antithrombin HI (human), factor IX (human), factor IX complex, plasma protein fractions.
  • the at least one thrombolytic enzyme can be at least one selected from alteplase, anistreplase, reteplase (recombinant), streptokinase, urokinase. (See, e.g., pp. 834-66 of Nursing 2001 Drug Handbook.)
  • the at least one alkylating drug can be at least one selected from busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cyclophosphamide, ifosfamide, lomustine, mechlorethamine hydrochloride, melphalan, melphalan hydrochloride, streptozocin, temozolomide, thiotepa.
  • the at least one antimetabolite can be at least one selected from capecitabine, cladribine, cytarabine, floxuridine, fludarabine phosphate, fluorouracil, hydroxyurea, mercaptopurine, methotrexate, methotrexate sodium, thioguanine.
  • the at least one antibiotic antineoplastic can be at least one selected from bleomycin sulfate, dactinomycin, daunorubicin citrate liposomal, daunorubicin hydrochloride, doxorubicin hydrochloride, doxorubicin hydrochloride liposomal, epirubicin hydrochloride, idarubicin hydrochloride, mitomycin, pentostatin, plicamycin, valrubicin.
  • the at least one antineoplastics that alter hormone balance can be at least one selected from anastrozole, bicalutamide, estramustine phosphate sodium, exemestane, flutamide, goserelin acetate, letrozole, leuprolide acetate, megestrol acetate, nilutamide, tamoxifen citrate, testolactone, toremifene citrate.
  • the at least one miscellaneous antineoplastic can be at least one selected from asparaginase, bacillus Calmette-Guerin (BCG) (live intravesical), dacarbazine, docetaxel, etoposide, etoposide phosphate, gemcitabine hydrochloride, irinotecan hydrochloride, mitotane, mitoxantrone hydrochloride, paclitaxel, pegaspargase, porfimer sodium, procarbazine hydrochloride, rituximab, teniposide, topotecan hydrochloride, trastuzumab, tretinoin, vinblastine sulfate, vincristine sulfate, vinorelbine tartrate.
  • BCG Bacillus Calmette-Guerin
  • the at least one immunosuppressant can be at least one selected from azathioprine, basiliximab, cyclosporine, daclizumab, lymphocyte immune globulin, muromonab-CD3, mycophenolate mofetil, mycophenolate mofetil hydrochloride, sirolimus, tacrolimus.
  • the at least one vaccine or toxoid can be at least one selected from BCG vaccine, cholera vaccine, diphtheria and tetanus toxoids (adsorbed), diphtheria and tetanus toxoids and acellular pertussis vaccine adsorbed, diphtheria and tetanus toxoids and whole-cell pertussis vaccine, Haemophilus b conjugate vaccines, hepatitis A vaccine (inactivated), hepatisis B vaccine (recombinant), influenza virus vaccine 1999-2000 trivalent types A & B (purified surface antigen), influenza virus vaccine 1999-2000 trivalent types A & B (subvirion or purified subvirion), influenza virus vaccine 1999-2000 trivalent types A & B (whole virion), Japanese encephalitis virus vaccine (inactivated), Lyme disease vaccine (recombinant OspA), measles and mumps and rubella virus vaccine (live), measles and mumps and rubella virus vaccine
  • the at least one antitoxin or antivenin can be at least one selected from black widow spider antivenin, Crotalidae antivenom (polyvalent), diphtheria antitoxin (equine), Micrurus fulvius antivenin).
  • the at least one immune serum can be at least one selected from cytomegalovirus immune globulin (intraveneous), hepatitis B immune globulin (human), immune globulin intramuscular, immune globulin intravenous, rabies immune globulin (human), respiratory syncytial virus immune globulin intravenous (human), Rho(D) immune globulin (human), Rho(D) immune globulin intravenous (human), tetanus immune globulin (human), varicella-zoster immune globulin.
  • cytomegalovirus immune globulin intraveneous
  • hepatitis B immune globulin human
  • immune globulin intramuscular immune globulin intravenous
  • rabies immune globulin human
  • respiratory syncytial virus immune globulin intravenous human
  • Rho(D) immune globulin human
  • Rho(D) immune globulin intravenous
  • the at least one biological response modifiers can be at least one selected from aldesleukin, GLP-letin alfa, filgrastim, glatiramer acetate for injection, interferon alfacon-1, interferon alfa-2a (recombinant), interferon alfa-2b (recombinant), interferon beta- Ia, interferon beta- Ib (recombinant), interferon gamma- Ib, levamisole hydrochloride, oprelvekin, sargramostim. (See, e.g., pp.
  • the at least one ophthalmic anti-infectives can be selected form bacitracin, chloramphenicol, ciprofloxacin hydrochloride, erythromycin, gentamicin sulfate, ofloxacin 0.3%, polymyxin B sulfate, sulfacetamide sodium 10%, sulfacetamide sodium 15%, sulfacetamide sodium 30%, tobramycin, vidarabine.
  • the at least one ophthalmic antiinflammatories can be at least one selected from dexamethasone, dexamethasone sodium phosphate, diclofenac sodium 0.1%, fluorometholone, flurbiprofen sodium, ketorolac tromethamine, prednisolone acetate (suspension) prednisolone sodium phosphate (solution).
  • the at least one miotic can be at least one selected from acetylcholine chloride, carbachol (intraocular), carbachol (topical), echothiophate iodide, pilocarpine, pilocarpine hydrochloride, pilocarpine nitrate.
  • the at least one mydriatic can be at least one selected from atropine sulfate, cyclopentplate hydrochloride, epinephrine hydrochloride, epinephryl borate, homatropine hydrobromide, phenylephrine hydrochloride, scopolamine hydrobromide, tropicamide.
  • the at least one ophthalmic vasoconstrictors can be at least one selected from naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride.
  • the at least one miscellaneous ophthalmics can be at least one selected from apraclonidine hydrochloride, betaxolol hydrochloride, brimonidine tartrate, carteolol hydrochloride, dipivefrin hydrochloride, dorzolamide hydrochloride, emedastine difumarate, fluorescein sodium, ketotifen fumarate, latanoprost, levobunolol hydrochloride, metipranolol hydrochloride, sodium chloride (hypertonic), timolol maleate.
  • the at least one otic can be at least one selected from boric acid, carbamide peroxide, chloramphenicol, triethanolamine polypeptide oleate-condensate.
  • the at least one nasal drug can be at least one selected from beclomethasone dipropionate, budesonide, ephedrine sulfate, epinephrine hydrochloride, flunisolide, fluticasone propionate, naphazoline hydrochloride, oxymetazoline hydrochloride, phenylephrine hydrochloride, tetrahydrozoline hydrochloride, triamcinolone acetonide, xylometazoline hydrochloride. (See, e.g., pp. 1041-97 of Nursing 2001 Drug Handbook)
  • the at least one local anti-infectives can be at least one selected from acyclovir, amphotericin B, azelaic acid cream, bacitracin, butoconazole nitrate, clindamycin phosphate, clotrimazole, econazole nitrate, erythromycin, gentamicin sulfate, ketoconazole, mafenide acetate, metronidazole (topical), miconazole nitrate, mupirocin, naftifine hydrochloride, neomycin sulfate, nitrofurazone, nystatin, silver sulfadiazine, terbinafine hydrochloride, terconazole, tetracycline hydrochloride, tioconazole, tolnaftate.
  • the at least one scabicide or pediculicide can be at least one selected from crotamiton, lindane, permethrin, and pyrethrins.
  • the at least one topical corticosteroid can be at least one selected from betamethasone dipropionate, betamethasone valerate, clobetasol propionate, desonide, desoximetasone, dexamethasone, dexamethasone sodium phosphate, diflorasone diacetate, fluocinolone acetonide, fluocinonide, flurandrenolide, fluticasone propionate, halcionide, hydrocortisone, hydrocortisone acetate, hydrocortisone butyrate, hydrocorisone valerate, mometasone furoate, triamcinolone acetonide. (See, e.g., pp. 1098-1136 of Nursing 2001 Drug Handbook.)
  • the at least one vitamin or mineral can be at least one selected from vitamin A, vitamin B complex, cyanocobalamin, folic acid, hydroxocobalamin, leucovorin calcium, niacin, niacinamide, pyridoxine hydrochloride, riboflavin, thiamine hydrochloride, vitamin C, vitamin D, cholecalciferol, ergocalciferol, vitamin D analogue, doxercalciferol, paricalcitol, vitamin E, vitamin K analogue, phytonadione, sodium fluoride, sodium fluoride (topical), trace elements, chromium, copper, iodine, manganese, selenium, zinc.
  • the at least one calorics can be at least one selected from amino acid infusions (crystalline), amino acid infusions in dextrose, amino acid infusions with electrolytes, amino acid infusions with electrolytes in dextrose, amino acid infusions for hepatic failure, amino acid infusions for high metabolic stress, amino acid infusions for renal failure, dextrose, fat emulsions, medium-chain triglycerides. (See, e.g., pp. 1137-63 of Nursing 2001 Drug Handbook.)
  • the present invention also provides at least one of any suitable and/or effective amount of a composition or pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant, optionally further comprise an effective amount of at least one further compound, protein or composition selected from at least one TNF antagonist (e.g., but not limited to a TNF chemical or protein antagonist, TNF monoclonal or polyclonal antibody or fragment, a soluble TNF receptor (e.g., p55, p70 or p85) or fragment, fusion polypeptides thereof, or a small molecule TNF antagonist, e.g., TNF binding protein I or II (TBP-I or TBP-II), nerelimonmab, infliximab, enteracept, CDP-571, CDP-870, afelimomab, lenercept, and the like), an antirheumatic (e.g., methotrexate, auranofin, aurothioglucose, a
  • GM-CSF GM-CSF, Leukine
  • an immunization an immunoglobulin, an immunosuppressive (e.g., basiliximab, cyclosporine, daclizumab), a growth hormone, a hormone replacement drug, an estrogen receptor modulator, a mydriatic, a cycloplegic, an alkylating agent, an antimetabolite, a mitotic inhibitor, a radiopharmaceutical, an antidepressant, antimanic agent, an antipsychotic, an anxiolytic, a hypnotic, a sympathomimetic, a stimulant, donepezil, tacrine, an asthma medication, a beta agonist, an inhaled steroid, a leukotriene inhibitor, a methylxanthine, a cromolyn, an epinephrine or analog, dornase alpha (Pulmozyme), a cytokine or a
  • Non- limiting examples of such cytokines include, but are not limted to, any of IL-I to IL-23. Suitable dosages are well known in the art. See, e.g., Wells et al., eds.,
  • compositions can also include toxin molecules that are associated, bound, co-formulated or co-administered with at least one antibody or polypeptide of the present invention.
  • the toxin can optionally act to selectively kill the pathologic cell or tissue.
  • the pathologic cell can be a cancer or other cell.
  • Such toxins can be, but are not limited to, purified or recombinant toxin or toxin fragment comprising at least one functional cytotoxic domain of toxin, e.g., selected from at least one of ricin, diphtheria toxin, a venom toxin, or a bacterial toxin.
  • the term toxin also includes both endotoxins and exotoxins produced by any naturally occurring, mutant or recombinant bacteria or viruses which may cause any pathological condition in humans and other mammals, including toxin shock, which can result in death.
  • Such toxins may include, but are not limited to, enterotoxigenic E.
  • coli heat-labile enterotoxin (LT), heat-stable enterotoxin (ST), Shigella cytotoxin, Aeromonas enterotoxins, toxic shock syndrome toxin-1 (TSST-I), Staphylococcal enterotoxin A (SEA), B (SEB), or C (SEC), Streptococcal enterotoxins and the like.
  • Such bacteria include, but are not limited to, strains of a species of enterotoxigenic E. coli (ETEC), enterohemorrhagic E.
  • coli e.g., strains of serotype 0157:H7
  • Staphylococcus species e.g., Staphylococcus aureus, Staphylococcus pyogenes
  • Shigella species e.g., Shigella dysenteriae, Shigella flexne ⁇ , Shigella boydii, and Shigella sonne ⁇
  • Salmonella species e.g., Salmonella typhi, Salmonella cholera-suis, Salmonella enteritidis
  • Clostridium species e.g., Clostridium perfringens, Clostridium perfringens, Clostridium perfringens, Clostridium pere, Clostridium botulinum
  • Camphlobacter species e.g., Camphlobacter jejuni, Camphlobacter fetus
  • Heliobacter species e.g., Heliobacter pylori
  • Aeromonas species
  • Aeromonas caviae Pleisomonas shigelloides, Yersina enterocolitica, Vibrios species (e.g., Vibrios cholerae, Vibrios parahemolyticus), Klebsiella species, Pseudomonas aeruginosa, and Streptococci.
  • Vibrios species e.g., Vibrios cholerae, Vibrios parahemolyticus
  • Klebsiella species Pseudomonas aeruginosa
  • Streptococci See, e.g., Stein, ed., INTERNAL MEDICINE, 3rd ed., pp 1-13, Little, Brown and Co., Boston, (1990); Evans et al., eds., Bacterial Infections of Humans: Epidemiology and Control, 2d.
  • GLP-I mimetibody or specified portion or variant compositions of the present invention can further comprise at least one of any suitable auxiliary, such as, but not limited to, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant or the like.
  • Pharmaceutically acceptable auxiliaries are preferred.
  • Non- limiting examples of, and methods of preparing such sterile solutions are well known in the art, such as, but limited to, Gennaro, Ed., Remington's Pharmaceutical Sciences, 18 th Edition, Mack Publishing Co. (Easton, PA) 1990.
  • Pharmaceutically acceptable carriers can be routinely selected that are suitable for the mode of administration, solubility and/or stability of the GLP-I mimetibody composition as well known in the art or as described herein.
  • Pharmaceutical excipients and additives useful in the present composition include but are not limited to proteins, peptides, amino acids, lipids, and carbohydrates (e.g., sugars, including monosaccharides, di-, tri ⁇ , tetra-, and oligosaccharides; derivatized sugars such as alditols, aldonic acids, esterified sugars and the like; and polysaccharides or sugar polymers), which can be present singly or in combination, comprising alone or in combination 1-99.99% by weight or volume.
  • Exemplary protein excipients include serum albumin such as human serum albumin (HSA), recombinant human albumin (rHA), gelatin, casein, and the like.
  • Representative amino acid/GLP-1 mimetibody or specified portion or variant components which can also function in a buffering capacity, include alanine, glycine, arginine, betaine, histidine, glutamic acid, aspartic acid, cysteine, lysine, leucine, isoleucine, valine, methionine, phenylalanine, aspartame, and the like.
  • One preferred amino acid is glycine.
  • Carbohydrate excipients suitable for use in the invention include, for example, monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol), myoinositol and the like.
  • Preferred carbohydrate excipients for use in the present invention are mannitol, trehalose, and raffinose.
  • GLP-I mimetibody compositions can also include a buffer or a pH adjusting agent; typically, the buffer is a salt prepared from an organic acid or base.
  • Representative buffers include organic acid salts such as salts of citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid, or phthalic acid; Tris, tromethamine hydrochloride, or phosphate buffers.
  • Preferred buffers for use in the present compositions are organic acid salts such as citrate.
  • the GLP-I mimetibody or specified portion or variant compositions of the invention can include polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl- ⁇ -cyclodextrin), polyethylene glycols, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antistatic agents, surfactants (e.g., polysorbates such as 'TWEEN 20" and "TWEEN 80"), lipids (e.g., phospholipids, fatty acids), steroids (e.g., cholesterol), and chelating agents (e.g., EDTA).
  • polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl- ⁇ -cyclod
  • the invention provides for stable formulations, which can preferably include a suitable buffer with saline or a chosen salt, as well as optional preserved solutions and formulations containing a preservative as well as multi- use preserved formulations suitable for pharmaceutical or veterinary use, comprising at least one GLP-I mimetibody or specified portion or variant in a pharmaceutically acceptable formulation.
  • Preserved formulations contain at least one known preservative or optionally selected from the group consisting of at least one phenol, m-cresol, p- cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (e.g., hexahydrate), alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent.
  • phenol m-cresol, p- cresol, o-cresol, chlorocresol
  • benzyl alcohol e.g., hexahydrate
  • alkylparaben methyl, ethyl, propyl, butyl and the like
  • Any suitable concentration or mixture can be used as known in the art, such as 0.001-5%, or any range or value therein, such as, but not limited to 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05, 0.09, 0.1, 0.2, 0.3, O.4., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2,
  • Non-limiting examples include, no preservative, 0.1-2% m-cresol (e.g., 0.2, 0.3.
  • benzyl alcohol e.g., 0.5, 0.9, 1.1., 1.5, 1.9, 2.0, 2.5%), 0.001-0.5% thimerosal (e.g., 0.005, 0.01), 0.001-2.0% phenol (e.g., 0.05, 0.25, 0.28, 0.5, 0.9, 1.0%), 0.0005-1.0% alkylparaben(s) (e.g., 0.00075, 0.0009, 0.001, 0.002, 0.005, 0.0075, 0.009, 0.01, 0.02, 0.05, 0.075, 0.09, 0.1, 0.2, 0.3, 0.5, 0.75, 0.9, 1.0%), and the like.
  • alkylparaben(s) e.g., 0.00075, 0.0009, 0.001, 0.002, 0.005, 0.0075, 0.009, 0.01, 0.02, 0.05, 0.075, 0.09, 0.1, 0.2, 0.3, 0.5, 0.75, 0.9, 1.0%),
  • the invention provides an article of manufacture, comprising packaging material and at least one vial comprising a solution of at least one GLP-I mimetibody or specified portion or variant with the prescribed buffers and/or preservatives, optionally in an aqueous diluent, wherein said packaging material comprises a label that indicates that such solution can be held over a period of 1, 2, 3, 4, 5, 6, 9, 12, 18, 20, 24, 30, 36, 40, 48, 54, 60, 66, 72 hours or greater.
  • the invention further comprises an article of manufacture, comprising packaging material, a first vial comprising lyophilized at least one GLP-I mimetibody or specified portion or variant, and a second vial comprising an aqueous diluent of prescribed buffer or preservative, wherein said packaging material comprises a label that instructs a patient to reconstitute the at least one GLP-I mimetibody or specified portion or variant in the aqueous diluent to form a solution that can be held over a period of twenty-four hours or greater.
  • the at least one GLP-I mimetibody or specified portion or variant used in accordance with the present invention can be produced by recombinant means, including from mammalian cell or transgenic preparations, or can be purified from other biological sources, as described herein or as known in the art.
  • the range of amounts of at least one GLP-I mimetibody or specified portion or variant in the product of the present invention includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 ⁇ g/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal patch, pulmonary, transmucosal, or osmotic or micro pump methods.
  • the aqueous diluent optionally further comprises a pharmaceutically acceptable preservative.
  • preservatives include those selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof.
  • concentration of preservative used in the formulation is a concentration sufficient to yield an anti-microbial effect. Such concentrations are dependent on the preservative selected and are readily determined by the skilled artisan.
  • excipients e.g. isotonicity agents, buffers, antioxidants, preservative enhancers
  • An isotonicity agent such as glycerin, is commonly used at known concentrations.
  • a physiologically tolerated buffer is preferably added to provide improved pH control.
  • the formulations can cover a wide range of pHs, such as from about pH 4 to about pH 10, and preferred ranges from about pH 5 to about pH 9, and a most preferred range of about 6.0 to about 8.0.
  • the formulations of the present invention have pH between about 6.8 and about 7.8.
  • Preferred buffers include phosphate buffers, most preferably sodium phosphate, particularly phosphate buffered saline (PBS).
  • additives such as a pharmaceutically acceptable solubilizers like Tween 20 (polyoxyethylene (20) sorbitan monolaurate), Tween 40 (polyoxyethylene (20) sorbitan monopalmitate), Tween 80 (polyoxyethylene (20) sorbitan monooleate), Pluronic F68 (polyoxyethylene polyoxypropylene block copolymers), and PEG (polyethylene glycol) or non-ionic surfactants such as polysorbate 20 or 80 or poloxamer 184 or 188, Pluronic® polyls, other block co-polymers, and chelators such as EDTA and EGTA can optionally be added to the formulations or compositions to reduce aggregation. These additives are particularly useful if a pump or plastic container is used to administer the formulation. The presence of pharmaceutically acceptable surfactant mitigates the propensity for the protein to aggregate.
  • a pharmaceutically acceptable solubilizers like Tween 20 (polyoxyethylene (20) sorbitan monolau
  • the formulations of the present invention can be prepared by a process which comprises mixing at least one GLP-I mimetibody or specified portion or variant and a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous diluent.
  • a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixture
  • aqueous diluent Mixing the at least one GLP-I mimetibody or specified portion or variant and preservative in an aqueous diluent is carried out using conventional dissolution and mixing procedures.
  • a suitable formulation for example, a measured amount of at least one GLP-I mimetibody or specified portion or variant in buffered solution is combined with the desired preservative in a buffered solution in quantities sufficient to provide the protein and preservative at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art. For example, the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that may be optimized for the concentration and means of administration used.
  • the claimed formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one GLP-I mimetibody or specified portion or variant that is reconstituted with a second vial containing water, a preservative and/or excipients, preferably a phosphate buffer and/or saline and a chosen salt, in an aqueous diluent.
  • a preservative and/or excipients preferably a phosphate buffer and/or saline and a chosen salt
  • compositions of the invention can optionally be safely stored at temperatures of from about 2 to about 40°C and retain the biologically activity of the protein for extended periods of time, thus, allowing a package label indicating that the solution can be held and/or used over a period of 6, 12, 18, 24, 36, 48, 72, or 96 hours or greater. If preserved diluent is used, such label can include use up to at least one of 1-12 months, one-half, one and a half, and/or two years.
  • the solutions of at least one GLP-I mimetibody or specified portion or variant in the invention can be prepared by a process that comprises mixing at least one GLP-I mimetibody or specified portion or variant in an aqueous diluent. Mixing is carried out using conventional dissolution and mixing procedures. To prepare a suitable diluent, for example, a measured amount of at least one GLP-I mimetibody or specified portion or variant in water or buffer is combined in quantities sufficient to provide the protein and optionally a preservative or buffer at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art. For example, the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that may be optimized for the concentration and means of administration used.
  • the claimed products can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one GLP-I mimetibody or specified portion or variant that is reconstituted with a second vial containing the aqueous diluent.
  • a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available.
  • the claimed products can be provided indirectly to patients by providing to pharmacies, clinics, or other such institutions and facilities, clear solutions or dual vials comprising a vial of lyophilized at least one GLP-I mimetibody or specified portion or variant that is reconstituted with a second vial containing the aqueous diluent.
  • the clear solution in this case can be up to one liter or even larger in size, providing a large reservoir from which smaller portions of the at least one GLP-I mimetibody or specified portion or variant solution can be retrieved one or multiple times for transfer into smaller vials and provided by the pharmacy or clinic to their customers and/or patients.
  • Recognized devices comprising these single vial systems include those pen- injector devices for delivery of a solution such as Humaject ® ' NovoPen ® , B-D ® Pen, AutoPen ® , and OptiPen ® .
  • Recognized devices comprising a dual vial system include those pen-injector systems for reconstituting a lyophilized drug in a cartridge for delivery of the reconstituted solution such as the HumatroPen " .
  • the products presently claimed include packaging material.
  • the packaging material provides, in addition to the information required by the regulatory agencies, the conditions under which the product can be used.
  • the packaging material of the present invention provides instructions to the patient to reconstitute the at least one GLP-I mimetibody or specified portion or variant in the aqueous diluent to form a solution and to use the solution over a period of 2-24 hours or greater for the two vial, wet/dry, product.
  • the label indicates that such solution can be used over a period of 2-24 hours or greater.
  • the presently claimed products are useful for human pharmaceutical product use.
  • the formulations of the present invention can be prepared by a process that comprises mixing at least one GLP-I mimetibody or specified portion or variant and a selected buffer, preferably a phosphate buffer containing saline or a chosen salt. Mixing the at least one GLP-I mimetibody or specified portion or variant and buffer in an aqueous diluent is carried out using conventional dissolution and mixing procedures. To prepare a suitable formulation, for example, a measured amount of at least one GLP- 1 mimetibody or specified portion or variant in water or buffer is combined with the desired buffering agent in water in quantities sufficient to provide the protein and buffer at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art.
  • the claimed stable or preserved formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one GLP-I mimetibody or specified portion or variant that is reconstituted with a second vial containing a preservative or buffer and excipients in an aqueous diluent.
  • a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available.
  • At least one GLP-I mimetibody or specified portion or variant in either the stable or preserved formulations or solutions described herein can be administered to a patient in accordance with the present invention via a variety of delivery methods including SC or IM injection; transdermal, pulmonary, transmucosal, implant, osmotic pump, cartridge, micro pump, or other means appreciated by the skilled artisan, as well- known in the art.
  • the present invention for mimetibodies also provides a method for modulating or treating diabetes, type I or type II diabetes mellitus, including adult onset or juvenile, insulin dependent, non-insulin dependent, and the like, including the associated signs and symptoms, such as but not limited to, insulin resistance, hyperglycemia, hypoglycemia, pancreatitis, Sushing's syndrome, acanthosis nigricans, lipoatrrophic diabetes, retinopathy, nephropathy, polyneuropathy, mononeuropathy, autonomic neuropathy, ulcers, foot ulcers, joint problems, infections (e.g., fungal or bacterial), and the like, in a cell, tissue, organ, animal, or patient.
  • diabetes type I or type II diabetes mellitus, including adult onset or juvenile, insulin dependent, non-insulin dependent, and the like, including the associated signs and symptoms, such as but not limited to, insulin resistance, hyperglycemia, hypoglycemia, pancreatitis, Sushing's
  • the present invention also provides a method for modulating or treating at least one diabetes associated immune related disease, in a cell, tissue, organ, animal, or patient including, but not limited to, at least one of type I or type II diabetes mellitus, including adult onset or juvenile, insulin dependent, non-insulin dependent, and the like, including the associated signs and symptoms, such as but not limited to, insulin resistance, hyperglycemia, hypoglycemia, pancreatitis, Sushing's syndrome, acanthosis nigricans, lipoatrrophic diabetes, retinopathy, nephropathy, polyneuropathy, mononeuropathy, autonomic neuropathy, ulcers, foot ulcers, joint problems, infections (e.g., fungal or bacterial), and the like.
  • type I or type II diabetes mellitus including adult onset or juvenile, insulin dependent, non-insulin dependent, and the like
  • associated signs and symptoms such as but not limited to, insulin resistance, hyperglycemia, hypoglycemia, pancrea
  • Such a method can optionally comprise administering an effective amount of at least one composition or pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • the present invention also provides a method for modulating or treating at least one cardiovascular disease in a cell, tissue, organ, animal, or patient, including, but not limited to, at least one of cardiac stun syndrome, myocardial infarction, congestive heart failure, stroke, ischemic stroke, hemorrhage, arteriosclerosis, atherosclerosis, diabetic ateriosclerotic disease, hypertension, arterial hypertension, renovascular hypertension, syncope, shock, syphilis of the cardiovascular system, heart failure, cor pulmonale, primary pulmonary hypertension, cardiac arrhythmias, atrial ectopic beats, atrial flutter, atrial fibrillation (sustained or paroxysmal), chaotic or multifocal atrial tachycardia, regular narrow QRS tachycardia, specific arrythmias, ventricular fibrillation, His bundle arrythmias, atrioventricular block, bundle branch block, myocardial ischemic disorders, coronary
  • Such a method can optionally comprise administering an effective amount of a composition or pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Any method of the present invention can comprise administering an effective amount of a composition or pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Such a method can optionally further comprise co-administration or combination therapy for treating such immune diseases, wherein the administering of said at least one GLP-I mimetibody, specified portion or variant thereof, further comprises administering, before concurrently, and/or after, at least one selected from at least one TNF antagonist (e.g., but not limited to a TNF antibody or fragment, a soluble TNF receptor or fragment, fusion proteins thereof, or a small molecule TNF antagonist), an antirheumatic, a muscle relaxant, a narcotic, a non- steroid anti-inflammatory drug (NSADD), an analgesic, an anesthetic, a sedative, a local anethetic, a neuromuscular blocker, an antimicrobial (e.g.,.
  • TNF antagonist e.g., but not limited to a TNF antibody or fragment, a soluble TNF receptor or fragment, fusion proteins thereof, or a small molecule TNF antagonist
  • an antirheumatic e.g.,
  • an antifungal an antiparasitic, an antiviral, a carbapenem, cephalosporin, a flurorquinolone, a macrolide, a penicillin, a sulfonamide, a tetracycline, another antimicrobial), an antipsoriatic, a corticosteriod, an anabolic steroid, a diabetes related agent, a mineral, a nutritional, a thyroid agent, a vitamin, a calcium related hormone, an antidiarrheal, an antitussive, an antiemetic, an antiulcer, a laxative, an anticoagulant, an erythropieitin (e.g., GLP-letin alpha), a filgrastim (e.g., G-CSF, Neupogen), a sargramostim (GM- CSF, Leukine), an immunization, an immunoglobulin, an immunosuppressive (e.g., basilixim
  • Suitable dosages are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2 nd Edition, Appleton and Lange, Stamford, CT (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, CA (2000), each of which references are entirely incorporated herein by reference.
  • Mimetibodies can also be used ex vivo, such as in autologous marrow culture. Briefly, bone marrow is removed from a patient prior to chemotherapy and treated with TPO and/or GLP-I, optionally in combination with mimetibodies, optionally in combination with one or more additional cytokines. The treated marrow is then returned to the patient after chemotherapy to speed the recovery of the marrow.
  • TPO alone and in combination with GLP-I mimetibodies and/or GLP-I, can also be used for the ex vivo expansion of marrow or peripheral blood progenitor (PBPC) cells.
  • PBPC peripheral blood progenitor
  • marrow Prior to chemotherapy treatment, marrow can be stimulated with stem cell factor (SCF) or G- CSF to release early progenitor cells into peripheral circulation.
  • SCF stem cell factor
  • G- CSF G- CSF
  • TPO tumor necrosis factor
  • mimetibodies optionally in combination with one or more other cytokines, including but not limited to SCF, G-CSF, IL-3, GM-CSF, IL-6 or IL-11, to differentiate and proliferate into high-density megakaryocyte cultures, which are optionally then be returned to the patient following high-dose chemotherapy.
  • Doses of TPO for ex vivo treatment of bone marrow will be in the range of 100 pg/ml to 10 ng/ml, preferably 500 pg/ml to 3 ng/ml. Doses of mimetibodies will be equivalent in activity to GLP . - 1 which can be used from 0.1 units/ml to 20 units/ml, preferably from 0.5 units/ml to 2 units/ml, or any range or value therein.
  • TNF antagonists suitable for compositions, combination therapy, coadministration, devices and/or methods of the present invention include, but are not limited to, anti-TNF antibodies, ligand-binding fragments thereof, and receptor molecules which bind specifically to TNF; compounds which prevent and/or inhibit TNF synthesis, TNF release or its action on target cells, such as thalidomide, tenidap, phosphodiesterase inhibitors (e.g, pentoxifylline and rolipram), A2b adenosine receptor agonists and A2b adenosine receptor enhancers; compounds which prevent and/or inhibit TNF receptor signalling, such as mitogen activated protein (MAP) kinase inhibitors; compounds which block and/or inhibit membrane TNF cleavage, such as metalloproteinase inhibitors; compounds which block and/or inhibit TNF activity, such as angiotensin converting enzyme (MAP) kinase inhibitors); compounds which block and/or inhibit membrane TNF cleavage,
  • a "tumor necrosis factor antibody,” “TNF antibody,” “TNF ⁇ antibody,” or fragment and the like decreases, blocks, inhibits, abrogates or interferes with TNF ⁇ activity in vitro, in situ and/or preferably in vivo.
  • a suitable TNF human antibody of the present invention can bind TNF ⁇ and includes anti-TNF antibodies, antigen-binding fragments thereof, and specified mutants or domains thereof that bind specifically to TNF ⁇ .
  • a suitable TNF antibody or fragment can also decrease block, abrogate, interfere, prevent and/or inhibit TNF RNA, DNA or protein synthesis, TNF release, TNF receptor signaling, membrane TNF cleavage, TNF activity, TNF production and/or synthesis.
  • Chimeric antibody cA2 consists of the antigen binding variable region of the high-affinity neutralizing mouse anti-human TNF ⁇ IgGl antibody, designated A2, and the constant regions of a human IgGl, kappa immunoglobulin.
  • the human IgGl Fc region improves allogeneic antibody effector function, increases the circulating serum half-life and decreases the immunogenicity of the antibody.
  • the avidity and epitope specificity of the chimeric antibody cA2 is derived from the variable region of the murine antibody A2.
  • a preferred source for nucleic acids encoding the variable region of the murine antibody A2 is the A2 hybridoma cell line.
  • Chimeric A2 (cA2) neutralizes the cytotoxic effect of both natural and recombinant human TNF ⁇ in a dose dependent manner. From binding assays of chimeric antibody cA2 and recombinant human TNF ⁇ , the affinity constant of chimeric antibody cA2 was calculated to be 1.04XlO 10 M "1 .
  • TNF Receptor Molecules Preferred TNF receptor molecules useful in the present invention are those that bind TNF ⁇ with high affinity (see, e.g., Feldmann et al, International Publication No. WO 92/07076 (published April 30, 1992); Schall et al, Cell 61:361-310 (1990); and Loetscher et al, Cell 67:351-359 (1990), which references are entirely incorporated herein by reference) and optionally possess low immunogenicity.
  • the 55 kDa (p55 TNF-R) and the 75 kDa (p75 TNF-R) TNF cell surface receptors are useful in the present invention.
  • Truncated forms of these receptors comprising the extracellular domains (ECD) of the receptors or functional portions thereof (see, e.g., Corcoran et al., Eur. J. Biochem. 223:831-840 (1994)), are also useful in the present invention.
  • Truncated forms of the TNF receptors, comprising the ECD have been detected in urine and serum as 30 kDa and 40 kDa TNF ⁇ inhibitory binding proteins (Engelmann, H. et al, J. Biol. Chem. 265:1531-1536 (1990)).
  • TNF receptor multimeric molecules and TNF immunoreceptor fusion molecules, and derivatives and fragments or portions thereof, are additional examples of TNF receptor molecules which are useful in the methods and compositions of the present invention.
  • the TNF receptor molecules which can be used in the invention are characterized by their ability to treat patients for extended periods with good to excellent alleviation of symptoms and low toxicity. Low immunogenicity and/or high affinity, as well as other undefined properties, may contribute to the therapeutic results achieved.
  • TNF receptor multimeric molecules useful in the present invention comprise all or a functional portion of the ECD of two or more TNF receptors linked via one or more polypeptide linkers or other nonpeptide linkers, such as polyethylene glycol (PEG).
  • the multimeric molecules can further comprise a signal peptide of a secreted protein to direct expression of the multimeric molecule.
  • TNF immunoreceptor fusion molecules useful in the methods and compositions of the present invention comprise at least one portion of one or more immunoglobulin molecules and all or a functional portion of one or more TNF receptors.
  • TNF immunoreceptor fusion molecules can be assembled as monomers, or hetero- or homo- multimers.
  • the immunoreceptor fusion molecules can also be monovalent or multivalent.
  • An example of such a TNF immunoreceptor fusion molecule is TNF receptor/IgG fusion protein.
  • TNF immunoreceptor fusion molecules and methods for their production have been described in the art (Lesslauer et al, Eur. J. Immunol. 2i:2883-2886 (1991); Ashkenazi et al., Proc. Natl. Acad. ScL USA 88:10535-10539 (1991); Peppel et al, J. Exp. Med. 774:1483-1489 (1991); Kolls et al, Proc.
  • a functional equivalent, derivative, fragment or region of TNF receptor molecule refers to the portion of the TNF receptor molecule, or the portion of the TNF receptor molecule sequence which encodes TNF receptor molecule, that is of sufficient size and sequences to functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNF ⁇ with high affinity and possess low immunogenicity).
  • a functional equivalent of TNF receptor molecule also includes modified TNF receptor molecules that functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNF ⁇ with high affinity and possess low immunogenicity).
  • a functional equivalent of TNF receptor molecule can contain a "SILENT" codon or one or more amino acid substitutions, deletions or additions (e.g., substitution of one acidic amino acid for another acidic amino acid; or substitution of one codon encoding the same or different hydrophobic amino acid for another codon encoding a hydrophobic amino acid).
  • SILENT substitution of one acidic amino acid for another acidic amino acid
  • substitution of one codon encoding the same or different hydrophobic amino acid for another codon encoding a hydrophobic amino acid See Ausubel et al, Current Protocols in Molecular Biology, Greene Publishing Assoc, and Wiley-Interscience, New York (1987-2003).
  • Cytokines include, but are not limited to all known cytokines. See, e.g., CopewithCytokines.com. Cytokine antagonists include, but are not limited to, any antibody, fragment or mimetic, any soluble receptor, fragment or mimetic, any small molecule antagonist, or any combination thereof.
  • Any method of the present invention can comprise a method for treating a protein mediated disorder, comprising administering an effective amount of a composition or pharmaceutical composition comprising at least one GLP-I mimetibody or specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Such a method can optionally further comprise coadministration or combination therapy for treating such immune diseases, wherein the administering of said at least one GLP-I mimetibody, specified portion or variant thereof, further comprises administering, before concurrently, and/or after, at least one selected from at least one other cytokines such as IL-3, -6 and -11; stem cell factor; G- CSF and GM-CSF.
  • the administering of said at least one GLP-I mimetibody, specified portion or variant thereof further comprises administering, before concurrently, and/or after, at least one selected from at least one other cytokines such as IL-3, -6 and -11; stem cell factor; G- CSF and GM-CSF.
  • treatment of pathologic conditions is effected by administering an effective amount or dosage of at least one GLP-I mimetibody composition that total, on average, a range from at least about 0.0001 to 500 milligrams of at least one GLP-I mimetibody or specified portion or variant /kilogram of patient per dose, and preferably from at least about 0.001 to 100 milligrams GLP-I mimetibody or specified portion or variant /kilogram of patient per single or multiple administration, depending upon the specific activity of contained in the composition.
  • the effective serum concentration can comprise 0.001-5000 ⁇ g/ml serum concentration per single or multiple adminstration.
  • Suitable dosages are known to medical practitioners and will, of course, depend upon the particular disease state, specific activity of the composition being administered, and the particular patient undergoing treatment. In some instances, to achieve the desired therapeutic amount, it can be necessary to provide for repeated administration, i.e., repeated individual administrations of a particular monitored or metered dose, where the individual administrations are repeated until the desired daily dose or effect is achieved.
  • Preferred doses can optionally include 0.0001, 0.0002, 0.0003, 0.0004, 0.0005. 0.0006, 0.0007, 0.0008, 00009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05.
  • the dosage administered can vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent, and its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired.
  • a dosage of active ingredient can be about 0.0001 to 100 milligrams per kilogram of body weight. Ordinarily 0.001 to 10, and preferably 0.001 to 1 milligrams per kilogram per administration or in sustained release form is effective to obtain desired results.
  • treatment of humans or animals can be provided as a one-time or periodic dosage of at least one GLP-I mimetibody or specified portion or variant of the present invention 0.0001 to 100 mg/kg, such as 0.0002, 0.0003, 0.0004, 0.0005. 0.0006, 0.0007, 0.0008, 00009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05.
  • Dosage forms (composition) suitable for internal administration generally contain from about 0.0001 milligram to about 500 milligrams of active ingredient per unit or container.
  • the active ingredient will ordinarily be present in an amount of about 0.5-99.999% by weight based on the total weight of the composition.
  • the GLP-I mimetibody or specified portion or variant can be formulated as a solution, suspension, emulsion or lyophilized powder in association, or separately provided, with a pharmaceutically acceptable parenteral vehicle.
  • a pharmaceutically acceptable parenteral vehicle examples include water, saline, Ringer's solution, dextrose solution, and 5% human serum albumin. Liposomes and nonaqueous vehicles such as fixed oils may also be used.
  • the vehicle or lyophilized powder may contain additives that maintain isotonicity (e.g., sodium chloride, mannitol) and chemical stability (e.g., buffers and preservatives).
  • the formulation is sterilized by known or suitable techniques.
  • Suitable pharmaceutical carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.
  • Therapeutic Administration Many known and developed modes of can be used for administering pharmaceutically effective amounts of at least one GLP-I mimetibody or specified portion or variant according to the present invention.
  • a GLP-I mimetibody of the present invention can be delivered in a carrier, as a solution, emulsion, colloid, or suspension, or as a powder, using any of a variety of devices and methods suitable for administration by inhalation or other modes described here within or known in the art.
  • Formulations for parenteral administration can contain as common excipients sterile water or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like.
  • Aqueous or oily suspensions for injection can be prepared by using an appropriate emulsifier or humidifier and a suspending agent, according to known methods.
  • Agents for injection can be a non-toxic, non-orally administrable diluting agent such as aquous solution or a sterile injectable solution or suspension in a solvent.
  • As the usable vehicle or solvent water, Ringer's solution, isotonic saline, etc.
  • sterile involatile oil can be used as an ordinary solvent, or suspending solvent.
  • any kind of involatile oil and fatty acid can be used, including natural or synthetic or semisynthetic fatty oils or fatty acids; natural or synthetic or semisynthtetic mono- or di- or tri-glycerides.
  • Parental administration is known in the art and includes, but is not limited to, conventional means of injections, a gas pressured needle-less injection device as described in U.S. Pat. No. 5,851,198, and a laser perforator device as described in U.S. Pat. No. 5,839,446 entirely incorporated herein by reference.
  • the invention further relates to the administration of at least one GLP-I mimetibody or specified portion or variant by parenteral, subcutaneous, intramuscular, intravenous, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal means.
  • Protein, GLP-I mimetibody or specified portion or variant compositions can be prepared for use for parenteral (subcutaneous, intramuscular or intravenous) administration particularly in the form of liquid solutions or suspensions; for use in vaginal or rectal administration particularly in semisolid forms such as creams and suppositories; for buccal, or sublingual administration particularly in the form of tablets or capsules; or intrahasally particularly in the form of powders, nasal drops or aerosols or certain agents; or transdermally particularly in the form of a gel, ointment, lotion, suspension or patch delivery system with chemical enhancers such as dimethyl sulfoxide to either modify the skin structure or to increase the drug concentration in the transdermal patch (Junginger, et al.
  • parenteral subcutaneous, intramuscular or intravenous
  • vaginal or rectal administration particularly in semisolid forms such as creams and suppositories
  • buccal, or sublingual administration particularly in the form of tablets or capsules
  • At least one GLP-I mimetibody or specified portion or variant composition is delivered in a particle size effective for reaching the lower airways of the lung or sinuses.
  • at least one GLP-I mimetibody or specified portion or variant can be delivered by any of a variety of inhalation or nasal devices known in the art for administration of a therapeutic agent by inhalation.
  • These devices capable of dGLP-1 siting aerosolized formulations in the sinus cavity or alveoli of a patient include metered dose inhalers, nebulizers, dry powder generators, sprayers, and the like.
  • GLP-I mimetibody or specified portion or variants are also known in the art. All such devices can use of formulations suitable for the administration for the dispensing of GLP-I mimetibody or specified portion or variant in an aerosol.
  • aerosols can be comprised of either solutions (both aqueous and non aqueous) or solid particles.
  • Metered dose inhalers like the Ventolin ® metered dose inhaler, typically use a propellent gas and require actuation during inspiration (See, e.g., WO 94/16970, WO 98/35888).
  • Dry powder inhalers like TurbuhalerTM (Astra), Rotahaler ® (Glaxo), Diskus ® (Glaxo), SpirosTM inhaler (Dura), devices marketed by Inhale Therapeutics, and the Spinhaler ® powder inhaler (Fisons), use breath-actuation of a mixed powder (US 4668218 Astra, EP 237507 Astra, WO 97/25086 Glaxo, WO 94/08552 Dura, US 5458135 Inhale, WO 94/06498 Fisons, entirely incorporated herein by reference).
  • Nebulizers like AERxTM Aradigm, the Ultravent ® nebulizer (Mallinckrodt), and the Acorn II ® nebulizer (Marquest Medical Products) (US 5404871 Aradigm, WO 97/22376), the above references entirely incorporated herein by reference, produce aerosols from solutions, while metered dose inhalers, dry powder inhalers, etc. generate small particle aerosols.
  • These specific examples of commercially available inhalation devices are intended to be a representative of specific devices suitable for the practice of this invention, and are not intended as limiting the scope of the invention.
  • a composition comprising at least one GLP-I mimetibody or specified portion or variant is delivered by a dry powder inhaler or a sprayer.
  • an inhalation device for administering at least one GLP-I mimetibody or specified portion or variant of the present invention.
  • delivery by the inhalation device is advantageously reliable, reproducible, and accurate.
  • the inhalation device can optionally deliver small dry particles, e.g. less than about 10 ⁇ m, preferably about 1-5 ⁇ m, for good respirability.
  • compositions as a Spray can be produced by forcing a suspension or solution of at least one GLP-I mimetibody or specified portion or variant through a nozzle under pressure.
  • the nozzle size and configuration, the applied pressure, and the liquid feed rate can be chosen to achieve the desired output and particle size.
  • An electrospray can be produced, for example, by an electric field in connection with a capillary or nozzle feed.
  • particles of at least one GLP-I mimetibody or specified portion or variant composition protein delivered by a sprayer have a particle size less than about 10 ⁇ m, preferably in the range of about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • Formulations of at least one GLP- 1 mimetibody or specified portion or variant composition protein suitable for use with a sprayer typically include GLP-I mimetibody or specified portion or variant composition protein in an aqueous solution at a concentration of about 1 mg to about 20 mg of at least one GLP-I mimetibody or specified portion or variant composition protein per ml of solution.
  • the formulation can include agents such as an excipient, a buffer, an isotonicity agent, a preservative, a surfactant, and, preferably, zinc.
  • the formulation can also include an excipient or agent for stabilization of the GLP-I mimetibody or specified portion or variant composition protein, such as a buffer, a reducing agent, a bulk protein, or a carbohydrate.
  • Bulk proteins useful in formulating GLP-I mimetibody or specified portion or variant composition proteins include albumin, protamine, or the like.
  • Typical carbohydrates useful in formulating GLP-I mimetibody or specified portion or variant composition proteins include sucrose, mannitol, lactose, trehalose, glucose, or the like.
  • the GLP-I mimetibody or specified portion or variant composition protein formulation can also include a surfactant, which can reduce or prevent surface-induced aggregation of the GLP-I mimetibody or specified portion or variant composition protein caused by atomization of the solution in forming an aerosol.
  • a surfactant which can reduce or prevent surface-induced aggregation of the GLP-I mimetibody or specified portion or variant composition protein caused by atomization of the solution in forming an aerosol.
  • Various conventional surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbitol fatty acid esters. Amounts will generally range between 0.001 and 14% by weight of the formulation.
  • Especially preferred surfactants for purposes of this invention are polyoxyethylene sorbitan monooleate, polysorbate 80, polysorbate 20, or the like. Additional agents known in the art for formulation of a protein such as mimetibodies, or specified portions or variants, can also be included in the formulation
  • GLP-I mimetibody or specified portion or variant compositions can be administered by a nebulizer, such as jet nebulizer or an ultrasonic nebulizer.
  • a nebulizer such as jet nebulizer or an ultrasonic nebulizer.
  • a compressed air source is used to create a high- velocity air jet through an orifice. As the gas expands beyond the nozzle, a low-pressure region is created, which draws a solution of GLP-I mimetibody or specified portion or variant composition protein through a capillary tube connected to a liquid reservoir.
  • the liquid stream from the capillary tube is sheared into unstable filaments and droplets as it exits the tube, creating the aerosol.
  • a range of configurations, flow rates, and baffle types can be employed to achieve the desired performance characteristics from a given jet nebulizer.
  • high- frequency electrical energy is used to create vibrational, mechanical energy, typically employing a piezoelectric transducer. This energy is transmitted to the formulation of GLP-I mimetibody or specified portion or variant composition protein either directly or through a coupling fluid, creating an aerosol including the GLP-I mimetibody or specified portion or variant composition protein.
  • particles of GLP-I mimetibody or specified portion or variant composition protein delivered by a nebulizer have a particle size less than about 10 ⁇ m, preferably in the range of about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • Formulations of at least one GLP-I mimetibody or specified portion or variant suitable for use with a nebulizer, either jet or ultrasonic typically include GLP-I mimetibody or specified portion or variant composition protein in an aqueous solution at a concentration of about 1 mg to about 20 mg of at least one GLP-I mimetibody or specified portion or variant protein per ml of solution.
  • the formulation can include agents such as an excipient, a buffer, an isotonicity agent, a preservative, a surfactant, and, preferably, zinc.
  • the formulation can also include an excipient or agent for stabilization of the at least one GLP-I mimetibody or specified portion or variant composition protein, such as a buffer, a reducing agent, a bulk protein, or a carbohydrate.
  • an excipient or agent for stabilization of the at least one GLP-I mimetibody or specified portion or variant composition protein such as a buffer, a reducing agent, a bulk protein, or a carbohydrate.
  • Bulk proteins useful in formulating at least one GLP-I mimetibody or specified portion or variant composition proteins include albumin, protamine, or the like.
  • Typical carbohydrates useful in formulating at least one GLP-I mimetibody or specified portion or variant include sucrose, mannitol, lactose, trehalose, glucose, or the like.
  • the at least one GLP-I mimetibody or specified portion or variant formulation can also include a surfactant, which can reduce or prevent surface-induced aggregation of the at least one GLP-I mimetibody or specified portion or variant caused by atomization of the solution in forming an aerosol.
  • a surfactant which can reduce or prevent surface-induced aggregation of the at least one GLP-I mimetibody or specified portion or variant caused by atomization of the solution in forming an aerosol.
  • Various conventional surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbital fatty acid esters. Amounts will generally range between 0.001 and 4% by weight of the formulation.
  • Especially preferred surfactants for purposes of this invention are polyoxyethylene sorbitan mono-oleate, polysorbate 80, polysorbate 20, or the like. Additional agents known in the art for formulation of a protein such as at least one GLP-I mimetibody or specified portion or variant protein can
  • a metered dose inhaler a propellant, at least one GLP-I mimetibody or specified portion or variant, and any excipients or other additives are contained in a canister as a mixture including a liquefied compressed gas. Actuation of the metering valve releases the mixture as an aerosol, preferably containing particles in the size range of less than about 10 ⁇ m, preferably about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • the desired aerosol particle size can be obtained by employing a formulation of GLP-I mimetibody or specified portion or variant composition protein produced by various methods known to those of skill in the art, including jet-milling, spray drying, critical point condensation, or the like.
  • Preferred metered dose inhalers include those manufactured by 3M or Glaxo and employing a hydrofluorocarbon propellant.
  • Formulations of at least one GLP-I mimetibody or specified portion or variant for use with a metered-dose inhaler device will generally include a finely divided powder containing at least one GLP-I mimetibody or specified portion or variant as a suspension in a non-aqueous medium, for example, suspended in a propellant with the aid of a surfactant.
  • the propellant can be any conventional material employed for this purpose, such as chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol and 1,1,1 ,2-tetrafluoroethane, HFA- 134a (hydrofluroalkane- 134a), HFA-227 (hydrofluroalkane-227), or the like.
  • the propellant is a . hydrofluorocarbon.
  • the surfactant can be chosen to stabilize the at least one GLP-I mimetibody or specified portion or variant as a suspension in the propellant, to protect the active agent against chemical degradation, and the like.
  • Suitable surfactants include sorbitan trioleate, soya lecithin, oleic acid, or the like. In some cases solution aerosols are preferred using solvents such as ethanol. Additional agents known in the art for formulation of a protein such as protein can also be included in the formulation.
  • compositions and methods of administering at least one GLP-I mimetibody or specified portion or variant include an emulsion comprising a plurality of submicron particles, a mucoadhesive macromolecule, a bioactive peptide, and an aqueous continuous phase, which promotes absorption through mucosal surfaces by achieving mucoadhesion of the emulsion particles (U.S. Pat. Nos. 5,514,670).
  • Mucous surfaces suitable for application of the emulsions of the present invention can include corneal, conjunctival, buccal, sublingual, nasal, vaginal, pulmonary, stomachic, intestinal, and rectal routes of administration.
  • Formulations for vaginal or rectal administration can contain as excipients, for example, polyalkyleneglycols, vaseline, cocoa butter, and the like.
  • Formulations for intranasal administration can be solid and contain as excipients, for example, lactose or can be aqueous or oily solutions of nasal drops.
  • excipients include sugars, calcium stearate, magnesium stearate, pregelinatined starch, and the like (U.S. Pat. Nos. 5,849,695).
  • Formulations for oral rely on the coadministration of adjuvants (e.g., resorcinols and nonionic surfactants such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether) to increase artificially the permeability of the intestinal walls, as well as the co-administration of enzymatic inhibitors (e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol) to inhibit enzymatic degradation.
  • adjuvants e.g., resorcinols and nonionic surfactants such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether
  • enzymatic inhibitors e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol
  • the active constituent compound of the solid-type dosage form for oral administration can be mixed with at least one additive, including sucrose, lactose, cellulose, mannitol, trehalose, raffinose, maltitol, dextran, starches, agar, arginates, chitins, chitosans, pectins, gum tragacanth, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride.
  • at least one additive including sucrose, lactose, cellulose, mannitol, trehalose, raffinose, maltitol, dextran, starches, agar, arginates, chitins, chitosans, pectins, gum tragacanth, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride.
  • These dosage forms can also contain other type(s) of additives, e.g., inactive diluting agent, lubricant such as magnesium stearate, paraben, preserving agent such as sorbic acid, ascorbic acid, alpha-tocopherol, antioxidant such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.
  • inactive diluting agent e.g., lubricant such as magnesium stearate, paraben
  • preserving agent such as sorbic acid, ascorbic acid, alpha-tocopherol, antioxidant such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.
  • antioxidant such as cysteine
  • Tablets and pills can be further processed into enteric-coated preparations.
  • the liquid preparations for oral administration include emulsion, syrup, elixir, suspension and solution preparations
  • Liposomes have also been described as drug delivery systems for insulin and heparin (U.S. Pat. No. 4,239,754). More recently, microspheres of artificial polymers of mixed amino acids (proteinoids) have been used to deliver pharmaceuticals (U.S. Pat. No. 4,925,673). Furthermore, carrier compounds described in U.S. Pat. No. 5,879,681 and U.S. Pat. No. 5,5,871,753 are used to deliver biologically active agents orally are known in the art.
  • the at least one GLP-I mimetibody or specified portion or variant is encapsulated in a delivery device such as a liposome or polymeric nanoparticles, microparticle, microcapsule, or microspheres (referred to collectively as microparticles unless otherwise stated).
  • a delivery device such as a liposome or polymeric nanoparticles, microparticle, microcapsule, or microspheres (referred to collectively as microparticles unless otherwise stated).
  • suitable devices are known, including microparticles made of synthetic polymers such as polyhydroxy acids such as polylactic acid, polyglycolic acid and copolymers thereof, polyorthoesters, polyanhydrides, and polyphosphazenes, and natural polymers such as collagen, polyamino acids, albumin and other proteins, alginate and other polysaccharides, and combinations thereof (U.S. Pat. Nos. 5,814,599).
  • a dosage form can contain a pharmaceutically acceptable non-toxic salt of the compounds that has a low degree of solubility in body fluids, for example, (a) an acid addition salt with a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene mono- or di- sulfonic acids, polygalacturonic acid, and the like; (b) a salt with a polyvalent metal cation such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed from e.g., N 9 N'- dibenzyl-ethylenediamine or ethylenediamine; or (c) combinations of (a) and (b) e.g.
  • a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, t
  • the compounds of the present invention or, preferably, a relatively insoluble salt such as those just described can be formulated in a gel, for example, an aluminum monostearate gel with, e.g. sesame oil, suitable for injection.
  • Particularly preferred salts are zinc salts, zinc tannate salts, pamoate salts, and the like.
  • Another type of slow release dGLP-lt formulation for injection would contain the compound or salt dispersed for encapsulated in a slow degrading, non-toxic, non- antigenic polymer such as a polylactic acid/polyglycolic acid polymer for example as described in U.S. Pat. No. 3,773,919.
  • the compounds or, preferably, relatively insoluble salts such as those described above can also be formulated in cholesterol matrix silastic pellets, particularly for use in animals.
  • Additional slow release, dGLP-lt or implant formulations, e.g. gas or liquid liposomes are known in the literature (U.S. Pat. Nos. 5,770,222 and "Sustained and Controlled Release Drug Delivery Systems",. J. R. Robinson ed., Marcel Dekker, Inc., N. Y., 1978).
  • Example 1 Cloning and Expression of a GLP-I mimetibody in Mammalian Cells.
  • a typical mammalian expression vector contains at least one promoter element, which mediates the initiation of transcription of mRNA, the GLP-I mimetibody or specified portion or variant coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Additional elements include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing.
  • LTRS long terminal repeats
  • CMV cytomegalovirus
  • cellular elements can also be used (e.g., the human actin promoter).
  • Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pIRESlneo, pRetro-Off, pRetro-On, PLXSN, or pLNCX (Clonetech Labs, Palo Alto, CA), pcDNA3.1 (+/-), pcDNA/Zeo (+/-) or pcDNA3.1/Hygro (+/-) (Invitrogen), PSVL and PMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109).
  • vectors such as pIRESlneo, pRetro-Off, pRetro-On, PLXSN, or pLNCX (Clonetech Labs, Palo Alto, CA), pcDNA3.1 (+/-), pcDNA/Zeo (+/-) or pcDNA3.1/Hy
  • Mammalian host cells that could be used include human HeIa 293, H9 and Jurkat cells, mouse NIH3T3 and C 127 cells, Cos 1, Cos 7 and CV 1, quail QC 1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.
  • the gene can be expressed in stable cell lines that contain the gene integrated into a chromosome.
  • a selectable marker such as dhfr, gpt, neomycin, or hygromycin allows the identification and isolation of the transfected cells.
  • the transfected gene can also be amplified to express large amounts of the encoded GLP-I mimetibody or specified portion or variant.
  • the DHFR (dihydrofolate reductase) marker is useful to develop cell lines that carry several hundred or even several thousand copies of the gene of interest.
  • Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy, et al., Biochem. J. 227:277-279 (1991); Bebbington, et al, Bio/Technology 10:169-175 (1992)). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified gene(s) integrated into a chromosome. Chinese hamster ovary (CHO) and NSO cells are often used for the production of GLP-I mimetibody or specified portion or variants.
  • GS glutamine synthase
  • the expression vectors pCl and pC4 contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen, et al., Molec. Cell. Biol. 5:438-447 (1985)) plus a fragment of the CMV-enhancer (Boshart, et al., Cell 41:521-530 (1985)). Multiple cloning sites, e.g., with the restriction enzyme cleavage sites BamHI, Xbal and Asp718, facilitate the cloning of the gene of interest.
  • the vectors contain in addition the 3' intron, the polyadenylation and termination signal of the rat preproinsulin gene.
  • Plasmid pC4 is a derivative of the plasmid pSV2-dhfr (ATCC Accession No. 37146).
  • the plasmid contains the mouse DHFR gene under control of the S V40 early promoter.
  • Chinese hamster ovary- or other cells lacking dihydrofolate activity that are transfected with these plasmids can be selected by growing the cells in a selective medium (e.g., alpha minus MEM, Life Technologies, Gaithersburg, MD) supplemented with the chemotherapeutic agent methotrexate.
  • MTX methotrexate
  • a second gene is linked to the DHFR gene, it is usually co-amplified and over-expressed. It is known in the art that this approach can be used to develop cell lines carrying more than 1,000 copies of the amplified gene(s). Subsequently, when the methotrexate is withdrawn, cell lines are obtained that contain the amplified gene integrated into one or more chromosome(s) of the host cell.
  • Plasmid pC4 contains for expressing the gene of interest the strong promoter of the long terminal repeat (LTR) of the Rous Sarcoma Virus (Cullen, et al., Molec. Cell. Biol. 5:438-447 (1985)) plus a fragment isolated from the enhancer of the immediate early gene of human cytomegalovirus (CMV) (Boshart, et al., Cell 41:521-530 (1985)). Downstream of the promoter are BamHI, Xbal, and Asp718 restriction enzyme cleavage sites that allow integration of the genes. Behind these cloning sites the plasmid contains the 3' intron and polyadenylation site of the rat preproinsulin gene.
  • LTR long terminal repeat
  • CMV cytomegalovirus
  • High efficiency promoters can also be used for the expression, e.g., the human b-actin promoter, the SV40 early or late promoters or the long terminal repeats from other retroviruses, e.g., HTV and HTLVI.
  • Clontech's Tet-Off and Tet-On gene expression systems and similar systems can be used to express the GLP-I in a regulated way in mammalian cells (M. Gossen, and H. Bujard, Proc. Natl. Acad. Sci. USA 89: 5547-5551 (1992)).
  • Other signals e.g., from the human growth hormone or globin genes can be used as well.
  • Stable cell lines carrying a gene of interest integrated into the chromosomes can also be selected upon co-transfection with a selectable marker such as gpt, G418 or hygromycin. It is advantageous to use more than one selectable marker in the beginning, e.g., G418 plus methotrexate.
  • the plasmid pC4 is digested with restriction enzymes and then dephosphorylated using calf intestinal phosphatase by procedures known in the art. The vector is then isolated from a 1% agarose gel.
  • the DNA sequence encoding the complete GLP-I mimetibody or specified portion or variant is used, corresponding to HC and LC variable regions of a GLP-I mimetibody of the present invention, according to known method steps.
  • Isolated nucleic acid encoding a suitable human constant region i.e., HC and LC regions is also used in this construct.
  • variable and constant region encoding DNA and the dephosphorylated vector are then ligated with T4 DNA ligase.
  • E. coli HBlOl or XL-I Blue cells are then transformed and bacteria are identified that contain the fragment inserted into plasmid pC4 using, for instance, restriction enzyme analysis.
  • CHO cells lacking an active DHFR gene are used for transfection. 5 ⁇ g of the expression plasmid pC4 is cotransfected with 0.5 ⁇ g of the plasmid pS V2-neo using lipofectin.
  • the plasmid pS V2neo contains a dominant selectable marker, the neo gene from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including G418.
  • the cells are seeded in alpha minus MEM supplemented with 1 ⁇ g /ml G418.
  • the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany) in alpha minus MEM supplemented with 10, 25, or 50 ng/ml of methotrexate plus 1 ⁇ g /ml G418. After about 10-14 days single clones are trypsinized and then seeded in 6- well petri dishes or 10 ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200 nM, 400 nM, 800 nM).
  • Clones growing at the highest concentrations of methotrexate are then transferred to new 6-well plates containing even higher concentrations of methotrexate (1 mM, 2 mM, 5 mM, 10 mM, 20 mM). The same procedure is repeated until clones are obtained that grow at a concentration of 100 - 200 mM. Expression of the desired gene product is analyzed, for instance, by SDS-PAGE and Western blot or by reverse phase HPLC analysis.
  • Example 2 Non-Limiting Example of a GLP-I mimetibody of the Invention.
  • GLP-I is a 37-amino acid peptide secreted from the L-cells of the intestine following an oral glucose challenge.
  • a mimetibody construct incorporating a biologically active GLP-I (7-37) peptide, variant or derivative is expected to prolong the in vivo lifetime of the peptide and provide a novel therapy for lowering blood glucose in Type 2 diabetic patients.
  • Peptides encoding the native GLP-I (7-37) peptide or a DPP- rV resistant analogue can be incorporated into the mimetibody scaffold.
  • the GLP-I peptide, the linker, the hinge, or the CH2 and CH3 sequences in the mimetibody could be deleted, added, substituted, mutated or modified to improve expression, potency, stability, or effector functions.
  • GLP-I GLP-I
  • A2S GLP-I
  • A2G GLP-I
  • A2G GLP-I
  • Mutations of the peptide could be made to improve the properties of a GLP-I mimetibody. For example mutations in the amino terminal residues may improve signaling while mutations in the helical domain may stabilize the helix and thereby improve binding to the receptor and/or stability of the mimetibody.
  • the length and composition of the linker could be mutated to vary the flexibility or stability of the attachment between the GLP-I peptide and the Fc region.
  • Different isotypes could be incorporated into the hinge region of the molecule.
  • mutations could be made within the hinge region of the mimetibody to stabilize the molecule.
  • the human IgG4 hinge could be mutated to make the Ser 228 - >Pro variant, to stabilize the interchain disulfide bonds in the mimetibody.
  • Variations within the Fc portion of the mimetibody could be made to improve the stability of the molecule and to change effector functions such as FcR binding.
  • one could use human or murine isotypes (or variations of these molecules) such as IgG4 with Ala/ Ala mutations.
  • GLP-I mimetibody of the Present Invention A specific, non-limiting, example of this invention is the GLP-I mimetibody construct (SEQ ID NO: 2) according to Formula (I):
  • P is a single copy of the bioactive GLP-I peptide (7-36)
  • L is a tandem repeat of either Gly-Ser or Gly-Gly-Gly-Ser flexible linker
  • V is the C-terminal of V H sequence, i.e., the J region of a naturally occurring IgG
  • H is the complete IgGl hinge region
  • CH2 & CH3 are of the IgGl isotype subclass. It is expected that the half-life of this construct will be many times that of the GLP-I peptide alone or its variant or derivative and similar to that of an IgG.
  • oligonucleotides comprised coding sequences for the GLP-I peptide, and a flexible linker composed of two GGGS repeats.
  • a restriction fragment containing the above-mentioned functional elements was then transferred into an expression vector.
  • This vector contained the anti-CD4 immunoglobulin promoter and enhancer, and the coding sequence for the human IgGl hinge . sequence, HC constant region 2 (CH2) and constant region 3 (CH3) as well as the necessary elements for plasmid replication and selection in bacteria and selection for stable expressers in mammalian cells.
  • This plasmid was introduced into the HEK293E cells and expression of the wt GLP-I MMB was achieved in transiently transfected cells. Purification of GLP-I MMB was accomplished by standard protein A and Superose 12 affinity chromatography, yielding approximately 1.5 mg/L of transfected cells. This protein was the starting material for the experiments described below.
  • GLP-I mimetibody The amino acid sequence of GLP-I mimetibody is shown in Figure 1. Functional domains are annotated above the peptide coding sequence. It is thought that the J sequence will provide even more flexibility to allow the GLP-I dimer to assume the proper conformation, and allow the dimer to protrude from the globular structure of the immunoglobulin and penetrate into the cleft between two GLP-I receptors. There are three cysteines in the IgGl hinge region. The first would normally pair to the immunoglobulin light chain (LC) and the other two participate in interchain bonds between two HCs. CH2 and CH3 regions constitute the bulk of the protein.
  • LC immunoglobulin light chain
  • GLP-I mimetibody contains two GLP-I peptides.
  • the spatial arrangement of the peptides at the N-terminus along with the flexibility of adjoining sequences should allow the peptides to form the bioacive dimer.
  • Example 3 FACS Binding Assay. The activity of GLP-I mimetibody was tested in an in vitro FACS binding assay. To determine whether the GLP-I MMB binds the GLP-IR, HEK293 cells (IxIO 6 cells) over-expressing the GLP-IR were incubated with GLP-I MMB (20 nM) for 2 hours at 4°C. The cells were washed, and a fluorescently labeled secondary detection antibody (1 ⁇ g/mL goat anti-human IgG, Fc gamma specific) was added for 30 minutes at 4°C. The fluorescence intensity of the cells was monitored via flow cytometry.
  • Figure 2 A shows that GLP-I MMB binds to HEK293 cells over-expressing the GLP-IR (grey, GLP-I MMB but no secondary; black, secondary only; red, negative control MMB and secondary; blue, GLP-I MMB and secondary).
  • Figure 2B shows that the GLP-I MMB does not bind to the control HEK293 cells (grey, GLP-I MMB but no secondary; black, secondary only; blue, GLP- 1 MMB and secondary).
  • FIG. 2C shows that a GLP-I peptide analogue (A2S) is able to compete with GLP-I MMB for binding to HEK293 cells over-expressing the GLP-IR (grey, GLP-I MMB but no secondary; black, GLP-I MMB and secondary; orange, GLP-I MMB, 0.2 nM competitor, secondary; blue, GLP-I MMB, 20 nM competitor, secondary; red, GLP-I MMB, 100 nM competitor, secondary).
  • A2S GLP-I peptide analogue
  • Example 5 DPP-IV cleavage assay. Since GLP-I is rapidly inactivated by DPP-IV, an in vitro assay was established to quantitate intact (i.e. uncleaved) GLP-I MMB. Briefly, GLP-I MMB or peptide (1.2 nM) was incubated at room temperature with DPP-IV (1 ⁇ g/mL, R&D Systems). After various times (0, 5, 10, 15, 20, 30, 40 minutes), a DPP-IV inhibitor (100 ⁇ M, Linco) was added to quench the reaction.
  • DPP-IV 1 ⁇ g/mL, R&D Systems
  • the amount of intact GLP-I MMB or peptide was measured using the GLP-I Active ELISA (Linco) and the GLP-I MMB or peptides for the respective standard curves.
  • Figure 4 shows that the GLP-I MMB was significantly more resistant to cleavage by DPP-IV, relative to the GLP-I peptide.
  • Example 6 Human Serum stability assay. The stability of the GLP-I MMB in serum was also measured to ensure that other serum proteases were not able to cleave and inactivate the GLP-I MMB. Briefly, GLPl peptide or the GLPl MMB (30 nM) was incubated in human serum at 37°C. After various times, the reactions were quenched with a DPP-IV inhibitor (100 ⁇ M, Linco), and the samples were analyzed using the GLP-I Active ELISA from Linco. Figure 5 shows that the GLP-I MMB is stable in human serum for 24 hours while the peptide is decayed rapidly.
  • Example 7 GLP-I MMB causes insulin secretion in RINm cells.
  • RINm cells were treated with increasing concentrations of GLP-I (7-36) peptide (0-5 nM), exendin-4 peptide (0-5 nM), or various GLP-I mimetibodies (5 or 50 nM) and the amount of insulin secreted was measured via ELISA. All GLP-I MMBs tested had activities in stimulating insulin secretion in RINm cells ( Figure 6). At 50 nM, the MMBs had activities comparable to that of the wide-type GLP-I (7-36) peptide.
  • Example 8 GLP-I MMB lowers glucose level in db/db mice.
  • Six week old db/db mice were fasted for two hours and then dosed intravenously with vehicle, GLP-I peptide, or GLP-I (A2S) MMB.
  • Blood glucose was monitored 0.5, 1, 2, 3, and 4 hours post-dosing.
  • the GLP-I peptide lowered blood glucose at 30 minutes, but by 60 minutes, the blood glucose began to increase again likely due to the short half-life of the GLP-I peptide.
  • GLP-I (A2S) MMB at a dose 100-fold lower than the GLP-I peptide dose induced a decrease in blood glucose throughout the entire 4 hour period (Figure 7A).
  • Example 9 Pharmacokinetics of GLP-I MMBs in mice and in cynomolgus monkeys.
  • A2G, A2S, exedin-cap, and wt mice were intravenously dosed with lmg/kg of the MMBs.
  • Plasma was obtained via cardiac puncture after sacrificing mice at different time point.
  • Various ELISAs were used to measure Fc, total MMB, active MMB, and acive peptide as they were metabolized in the animal.
  • Active MMB reflects the intact N-terminus of the peptide still attached to the Fc region of the mimetibody. Substitution of the second amino acid in the peptide (alanine) with either a serine or a glycine prolonged the lifetime of the active MMB in circulation.
  • Example 11 Effects of GLP-I MMB on fasting blood glucose during chronic dosing to diabetic mice.
  • Ten-week old diabetic db/db mice were subcutaneously dosed daily with vehicle or GLP-I MMB (1 mg/kg) for six weeks.
  • Fasting blood glucose was measured twice per week during the course of the study.
  • the fasting blood glucose was reduced in the treated animals relative to the controls throughout the study ( Figure 10), and by six weeks, the difference was more than 200 mg/dL (466 vs 221 mg/dL, control and treated animals respectively).
  • Example 12 Effects of GLP-I MMB on oral glucose tolerance test after chronic dosing to diabetic mice.
  • Example 13 Effects of GLP-I MMB on reducing HbAIc after chronic dosing to diabetic mice.
  • ten- week old diabetic db/db mice were dosed daily with vehicle or GLP-I MMB (1 mg/kg) for six weeks.
  • whole blood samples were taken and analyzed for percent HbAIc.
  • the HbAlC of the GLP-I treated animals increased by 109 percent during the six-week period whereas the control treated animals increased by 142 percent.
  • Example 14 Effects of GLP-I MMB on an oral glucose tolerance test in normal cynomolgus monkeys.
  • Example 15 Effects of GLP-I MMB on insulin staining in islets of diabetic mice (db/db) after a single dose. Twelve-week old diabetic mice (db/db) were treated with a single subcutaneous dose of the GLP-I MMB (1.5 mg/kg), and the pancreata were harvested four weeks later. The pancreata were sectioned and stained for the presence of insulin. As shown in Figure 14, there was significantly more insulin staining in the treated animals relative to the control animals.
  • Example 16 GLP-I MMB delays gastric emptying in normal dogs.
  • a gastric cannula was surgically implanted into female beagle dogs (10-15 kg) under general anesthesia and allowed to recover for at least 2 weeks. Dogs were fasted for 24 hours after which water was freely available. The gastric cannula was opened and gastric juice and food remnants were removed with 40-50 ml of lukewarm water. Groups of six dogs were dosed subcutaneously with lidamidine, an alpha2 agonist (0.63 mg/kg), 60 minutes before the meal, a positive control for delay of gastric emptying.
  • Dogs dosed with the vehicle control or GLP-I MMB (0.1 mg/kg) were dosed intravenously in the cephalic vein 15 minutes before the meal.
  • the gastric cannula was opened to determine the amount of fluid present in stomach for baseline value and fluid was promptly reintroduced.
  • a test meal consisting of 250 ml of a glucose solution (5 g/1) was administered via the cannula and allowed to remain in the stomach for 30 minutes. Gastric contents were drained from the stomach to measure total volume after 30 minutes. One ml of gastric contents was retained for analysis and the remaining volume was reintroduced into the stomach via the cannula.
  • Example 17 GLP-I MMB lowers blood glucose following an oral glucose tolerance test (OGTT) in diet induced obese mice.
  • OGTT oral glucose tolerance test
  • mice were maintained on a high fat diet for at least 27 weeks. Mice became obese and were determined to be diabetic when fasting blood glucose values exceeded 120 mg/dl.
  • diet induced obese mice were fasted overnight and dosed subcutaneously with 0.02, 0.2, or 2 mg/kg GLP-I MMB or vehicle control. Six hours after dosing, mice were given a 1.5 mg/g gastric gavage of glucose.
  • Example 18 GLP-I MMB lowers blood glucose in an intraperitoneal glucose tolerance test (IPGTT) in db/db mice. Male db/db mice of approximately 13- 15 weeks of age were randomized into treatment groups of six mice based on fasting blood glucose levels.
  • IPGTT intraperitoneal glucose tolerance test
  • Example 19 GLP-I MMB Dose-Dependently Inhibits Cytokine-Induced Apoptosis.
  • RIN-m cells were seeded at 50,000 cells/well in 96 well plates and incubated at 37 0 C overnight. The following day, cells were treated with a dose range of GLP-I MMB (serially diluted from 10OnM) for 30 minutes before the addition of the cytokines TNF ⁇ (lOng/mL) or IL-l ⁇ (4ng/mL). The plates were incubated at 37°C for 16 hours. To assay apoptosis the Cell Death ELISA-Plus assay kit (Roche Applied Science, Cat. No. 11920685001) was used.
  • ⁇ -cell apoptosis may play a role in diabetes development in two distinct phases of the disease.
  • developmental wave of islet apoptosis may affect the T-cell biology that ultimately impact on the onset of autoimmune diabetes.
  • islet apoptosis is also the mode of cell death that finally results in the massive destruction of ⁇ -cells.
  • the use of the GLP-I MMB to inhibit ⁇ -cell apoptosis could therefore be useful in the prevention of the autoimmune onset of diabetes or the progression onto overt diabetes by preserving ⁇ -cell mass.
  • TlD is a result from an irreversible loss of insulin-secreting beta cells.
  • insulin secretion is detectable in some people with long-standing TlD, indicating either a small population of surviving beta cells or continued renewal of beta cells subject to ongoing autoimmune destruction. It has been shown that TlD have ⁇ - cells that continually undergo apoptosis following replication. Beta cell apoptosis is twice as frequent in TlD as in control subjects. Most people with long-standing TlD have beta cells that continue to be destroyed. The mechanisms underlying increased beta cell death may involve both ongoing autoimmunity and glucose toxicity.
  • TlD may be prevented or reversed by targeted GLP-I MMB inhibition of ⁇ -cell destruction.
  • islets in the pancreas are subject to apoptosis and necrosis during the events that lead to brain death for cadaveric organ donors that continue during their clinical course prior to organ donation.
  • islets are subject to apoptosis during the islet isolation process in preparation of the insulin- producing cells for implantation.
  • cytokine activation specifically TNF ⁇ , ILlB, and ⁇ lNF.
  • the use of GLP -1 MMB in islet transplantation in the donor, during the isolation process, and during and following transplantation reduces the apoptosis and improve islet function. It also reduces the number of islets required for clinical benefit and provides an opportunity for the use of living donors in islet transplantation.
  • Example 20 GLP-I MMB Increases Glucose-Dependent Insulin Secretion in INS-IE Cells.
  • BSfS-IE cells were plated in 24-well plates at a cell density of 200,000 cells/well in RPMI 1640 + 10% FBS + 1% L-glutamine + 1% Sodium Pyruvate + 1% Non-essential Amino Acids + 50 ⁇ M ⁇ -Mercaptoethanol media. The cells were allowed to grow for 7 days at 37°C and 5% CO 2 and were fed on day 3 or 4. The cells were washed twice with 0.4 ml of KRBH buffer/3 mM glucose and allowed to incubate in this buffer for 30-60 minutes.
  • Example 21 GLP-I MMB Increases Glucose-Dependent Insulin Secretion in Rat and Human Islets.
  • Rat islets were obtained by collagenase digestion followed by Ficoll gradient purification. Human islets were isolated using an enzymatic digestion and density gradient purification described in detail in Ricordi et al. Diabetes (1998) 37; 4, 413-420. Islets were plated overnight in CMRL-1066 (Gibco), pen/strep (5,000units/5,000 ⁇ g), 10%FBS, 1% L-Glutamine 25 mM HEPES, pH 7.2-7.4 media in a 20 x 100 mm petri dish, and cultured at 37°C in 5% CO 2 for 18 to 24 hours.
  • islets were swirled to center of dish and collected into a 50 mL conical tube.
  • the islets were allowed to settle to the bottom of the tube by gravity for approximately 10-15 minutes.
  • the supernatant was removed, Functionality/ Viability media (MediaTech, cat#99-786-CV) was added and islets were allowed to settle to the bottom of the tube by gravity.
  • the islets were washed for a total of three times.
  • the supernatant was removed and the islets were resuspended at a density of 20 islets per mL in Functionality/ Viability media containing 1%BSA, 1% L-Glutamine, 1% pen/strep and 0.5 mM glucose solution.
  • Rat insulin was quantitated using the Crystal Chem (Downers Grove, IL) Ultra-Sensitive ELISA Assay kit (cat # 90060). Human insulin was quantitated using the Human Insulin ELISA kit from Linco Research (St. Charles, MI, cat#EZHI-14K).
  • Figure 20 shows that GLP-I MMB significantly increases insulin secretion from rat ( Figure 20A) and human islets (Figure 20B).
  • Diabetes both type 1 and type 2 is ultimately due to an insufficient amount of insulin being secreted by an individual's ⁇ -cells.
  • the ability to enhance the insulin secretion of individuals with diabetes by treating them with GLP-I MMB would improve or eliminate their diabetic condition.
  • the increase in insulin secretion is glucose dependant, only increasing in the presence of elevated glucose levels. Therefore, the risk of a patient experiencing dangerous hypoglycemic episodes is minimized with the use of the GLP-I MMB.
  • an increase in insulin secretion would also benefit an islet transplantation procedure. By increasing the insulin secreting capacity of transplanted islets, one could effectively reduce the number of islets required to provide clinical benefit.
  • Example 22 The effects of GLP-I MMB on the blood glucose of normal mice. Normal C57BLK/6 mice were randomized into two groups with each group having 13 animals. Group 1 received daily intraperitoneal (IP) doses of PBS vehicle or GLP-I MMB (0.5 mg/kg).
  • IP intraperitoneal
  • mice were treated for 10 days. The mice were monitored daily for body weight and blood glucose was monitored (LifeScan, CA) for 32 days at which point the study was terminated. [274] Mice receiving GLP-I MMB treatment showed lower blood glucose during the course of the 10-day treatment as compared to the PBS treated mice ( Figure 21). Blood glucose returned to the level of the PBS group once treatment was terminated. Both groups showed identical body weight gain throughout the course of the study (data not shown).
  • OGTT oral glucose tolerance test
  • Morphometric analysis of insulin staining showed a significantly higher intensity of insulin staining in GLP-I MMB treated mice versus controls. The experiment was repeated with the mice sacrificed 14 days after dosing to evaluate the longevity of the single dose of GLP-I MMB. Again, morphometric analysis of the insulin staining showed a higher intensity of insulin staining in the GLP-I MMB treated animals two weeks following a single dose. The intensity of insulin staining is directly related to the amount of stored insulin within the islets suggesting that GLP-I MMB treatment increases insulin secretion and storage in the islets of this diabetic animal model.
  • Example 24 The effects of GLP-I MMB on the efficacy of a marginal mass transplant of human islets in diabetic immunocompromised mice.
  • Athymic nu/nu (nude) mice were purchased from Harlan Laboratories. Animals were housed in Virus Antibody Free rooms in micro-isolated cages with free access to sterilized food and water. All experiments were conducted in accordance with the standards set forth in the Guide for the Care and Use of Laboratory Animals.
  • Diabetes was induced by intravenous administration of streptozotocin
  • GLP-I MMB treated mice achieved normoglycemia more rapidly relative to the control mice following the transplant of a marginal mass of islets.
  • GLP-I MMB treated mice demonstrated a more efficient clearance of a glucose challenge.
  • Example 25 The effects of GLP-I MMB on the insulin secretion in response to glucose in nonhuman primate islets.
  • Nonhuman primate islets were isolated using an enzymatic digestion and density gradient purification and were divided into two groups and cultured at 37°C in humidified mixed 95%air/5% CO 2 in non-tissue culture treated 175 cm 2 flasks (Corning, MA) at a density of ⁇ 20,000 IE.
  • One group of islets was cultured alone and the other group was cultured in MMl supplemented with 50 nM GLP-I MMB.
  • Islets were collected and a sample of islets (2 from control and 1 from GLP-I MMB groups) was subjected to a dynamic stimulation assay to evaluate the effects of glucose concentration on the insulin secretion of the islets. Islets were pre-perifused in a chromatography column (Bio-gel Fine 45-90 nm;
  • Example 26 The effects of GLP-I MMB on allogeneic islet engraftment and long-term survival in a cynomolgus monkey marginal mass model.
  • the use of a non-human primate marginal mass model enables one to study transplant approaches that have the potential to allow for reversal of diabetes with a reduced number of islets.
  • the marginal mass model approximately 5,000 IEQ/kg, or half the number (usually 10,000 IEQ/kg) required to achieve normoglycemia, are transplanted.
  • islets obtained from the same allogeneic donor one eliminates donor and isolation variables.
  • transplanting pairs of monkeys that have similar pre-transplant insulin requirements it is possible to observe enhanced islet engraftment in the monkey treated with an agent designed to enhance islet function and limit early islet loss as compared to the monkey that does not receive the agent.
  • Recipients were 2-3 year old cynomolgus monkeys of Mauritian origin; donors of the same strain were > 4 years of age. Diabetes was induced with 1250 mg/m 2 streptozotocin. Diabetic animals were treated with NPH insulin before the morning meal and with NPH/Lantus before the afternoon meal in an attempt to maintain blood glucose levels between 150-300 mg/dl. Four weeks after diabetes induction, the monkeys underwent a glucagon challenge to verify that no stimulated c-peptide was produced.
  • the donor organ was harvested and islets isolated via the semi-automated method referenced in Example A and Example B, followed by 2 overnight culture periods, first at 37 0 C and then at 22 0 C.
  • Islets for the control monkey were cultured in standard MMl media and islets for the experimental monkeys were cultured in MMl supplemented with 50 nM GLP-I MMB.
  • Monkeys underwent a mini-laparotomy and islet transplantation was performed via the portal vein into the liver.
  • a pair of monkeys received ⁇ 5,000 IEQ/kg from the same donor (cultured as described above); one monkey received GLP-I MMB (0.2 mg/kg) subcutaneously two times per week and the control monkey did not.
  • SFIS steroid free immune suppression
  • FK506, high dose rapamycin, anti-IL2R induction therapy steroid free immune suppression
  • Animals were monitored twice a day to determine fasting and post-prandial blood glucose levels (heel stick, glucometer) and treated with insulin as needed to maintain blood glucose in the 100-200 mg/dl range post-transplant.
  • Fasting c-peptide was determined every other week and intravenous glucose tolerance testing (IVGTT) was done every 8 weeks to assess graft function.
  • GLP-I MMB treated animal had superior glucose control with a significant reduction in exogenous insulin requirements (Figure 24A).
  • the improved glucose control was further illustrated by the rapid reduction in the HgbAlc in the GLP-I MMB treated animal as compared to the non-treated control ( Figure 24B). This study further supports the utilization of GLP-I MMB in a type 1 diabetic undergoing an islet transplant procedure.
  • GLP-I analogues The use of this novel molecule as a therapeutic to treat type 2 diabetes provides several advantages over other GLP-I analogues. For example, it is likely to prolong the half-life of the GLP-I peptide. Also, the wild-type GLP-I peptide in the mimetibody scaffold is resistant to protease degradation, specifically DPP-IV. This may allow for treatment with the wild-type GLP-I peptide rather than a mutant peptide. Since GLP-I is a native peptide, there may be less immune response in patients treated with a GLP-I mimetibody than in patients treated with a mutated GLP-I analogue.
  • the large size of the GLP-I MMB may preclude it from crossing the blood brain barrier. This may offer an advantage since nausea and anxiety have been associated with GLP-I engaging the GLP-IR in the brain.
  • the mimetibody platform results in expression of two peptides on each mimetibody molecule. This may allow the GLP-I peptides to interact with each other, forming a dimeric ligand that could increase affinity to the cell surface GLP-I receptor.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Endocrinology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Cardiology (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne au moins un nouveau miméticorps de la GLP-1 humaine ou une partie ou un variant spécifié, comprenant des acides nucléiques isolés qui codent pour au moins un miméticorps de la GLP-1 ou une partie ou un variant spécifié, un miméticorps de la GLP-1 ou une partie ou des variants spécifiés, des vecteurs, des cellules hôtes, des animaux ou des végétaux transgéniques, ainsi que des procédés pour les produire et les utiliser, y compris des compositions, des procédés et des dispositifs thérapeutiques.
EP05858723A 2005-03-28 2005-12-22 Mimeticorps de la glp-1 humaine, compositions, procedes et utilisations Withdrawn EP1871811A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US2005097175 2005-03-28
PCT/US2005/046884 WO2007081302A2 (fr) 2005-03-28 2005-12-22 Miméticorps de la glp-1 humaine, compositions, procédés et utilisations

Publications (2)

Publication Number Publication Date
EP1871811A2 true EP1871811A2 (fr) 2008-01-02
EP1871811A4 EP1871811A4 (fr) 2009-07-22

Family

ID=38256744

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05858723A Withdrawn EP1871811A4 (fr) 2005-03-28 2005-12-22 Mimeticorps de la glp-1 humaine, compositions, procedes et utilisations

Country Status (12)

Country Link
EP (1) EP1871811A4 (fr)
JP (1) JP2008546373A (fr)
KR (1) KR20080005378A (fr)
CN (1) CN101389346A (fr)
AU (1) AU2005339797A1 (fr)
BR (1) BRPI0520168A2 (fr)
CA (1) CA2603359A1 (fr)
CR (1) CR9481A (fr)
IL (1) IL186307A0 (fr)
MX (1) MX2007011975A (fr)
NO (1) NO20075272L (fr)
WO (1) WO2007081302A2 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR064623A1 (es) 2006-12-21 2009-04-15 Centocor Inc Uso de agonistas del receptor de glp -1 de accion prolongada para mejorar la sensibilidad a la insulina y los perfiles lipidicos
CN111574583B (zh) * 2020-04-10 2021-06-25 上海海路生物技术有限公司 蛋白复性试剂及其应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002046227A2 (fr) * 2000-12-07 2002-06-13 Eli Lilly And Company Proteines hybrides glp-1
WO2004002424A2 (fr) * 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques d'epo de mammifere a deletion ch1, compositions, methodes et utilisations associees
WO2005005604A2 (fr) * 2003-06-30 2005-01-20 Centocor, Inc. Proteines derivees de l'immunoglobuline anti-cible et genetiquement modifiees, compositions, procedes et utilisations
WO2005097175A2 (fr) * 2004-03-31 2005-10-20 Centocor, Inc. Corps mimetiques glp-1 humains, compositions, procedes et utilisations

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3224700A (en) * 1999-02-08 2000-08-25 Chiron Corporation Fibroblast growth factor receptor-immunoglobulin fusion
WO2004002417A2 (fr) * 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques mammaliens a deletion ch1, compositions, procedes et utilisations

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002046227A2 (fr) * 2000-12-07 2002-06-13 Eli Lilly And Company Proteines hybrides glp-1
WO2004002424A2 (fr) * 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques d'epo de mammifere a deletion ch1, compositions, methodes et utilisations associees
WO2005005604A2 (fr) * 2003-06-30 2005-01-20 Centocor, Inc. Proteines derivees de l'immunoglobuline anti-cible et genetiquement modifiees, compositions, procedes et utilisations
WO2005097175A2 (fr) * 2004-03-31 2005-10-20 Centocor, Inc. Corps mimetiques glp-1 humains, compositions, procedes et utilisations

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ADELHORST K ET AL: "STRUCTURE-ACTIVITY STUDIES OF GLUCAGON-LIKE PEPTIDE-1" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOCHEMICAL BIOLOGISTS, BIRMINGHAM, US, vol. 269, no. 9, 4 March 1994 (1994-03-04), pages 6275-6278, XP002045291 ISSN: 0021-9258 *
See also references of WO2007081302A2 *

Also Published As

Publication number Publication date
NO20075272L (no) 2007-12-28
AU2005339797A1 (en) 2007-07-19
WO2007081302A3 (fr) 2008-11-13
JP2008546373A (ja) 2008-12-25
CR9481A (es) 2009-01-14
CN101389346A (zh) 2009-03-18
CA2603359A1 (fr) 2007-07-19
KR20080005378A (ko) 2008-01-11
EP1871811A4 (fr) 2009-07-22
MX2007011975A (es) 2008-03-14
IL186307A0 (en) 2008-01-20
WO2007081302A2 (fr) 2007-07-19
BRPI0520168A2 (pt) 2009-04-22

Similar Documents

Publication Publication Date Title
US8071103B2 (en) Pharmaceutical composition comprising a human GLP-1 mimetibody
US20070128193A1 (en) GLP-1 agonists, compositions, methods and uses
US20070135338A1 (en) Human GLP-1 mimetibodies, compositions, methods and uses
AU2004277884B2 (en) Human EPO mimetic hinge core mimetibodies, compositions, methods and uses
US7241733B2 (en) Mammalian EPO mimetic CH1 deleted mimetibodies, compositions, methods and uses
US20100249032A1 (en) Human EPO Mimetic Hinge Core Mimetibodies, Compositions, Methods and Uses
CA2673657A1 (fr) Utilisation d'agonistes du recepteur glp-1 a action prolongee pour ameliorer la sensibilite a l'insuline et les profils des lipides
WO2007081302A2 (fr) Miméticorps de la glp-1 humaine, compositions, procédés et utilisations
AU2011202563A1 (en) Human EPO mimetic hinge core mimetibodies, compositions, methods and uses

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071017

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

R17D Deferred search report published (corrected)

Effective date: 20081113

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/00 20060101ALI20081125BHEP

Ipc: A61K 38/00 20060101AFI20081125BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20090618

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CENTOCOR ORTHO BIOTECH INC.

17Q First examination report despatched

Effective date: 20091007

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

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

18D Application deemed to be withdrawn

Effective date: 20100420