MXPA06003677A - Human epo mimetic hinge core mimetibodies, compositions, methods and uses - Google Patents

Human epo mimetic hinge core mimetibodies, compositions, methods and uses

Info

Publication number
MXPA06003677A
MXPA06003677A MXPA/A/2006/003677A MXPA06003677A MXPA06003677A MX PA06003677 A MXPA06003677 A MX PA06003677A MX PA06003677 A MXPA06003677 A MX PA06003677A MX PA06003677 A MXPA06003677 A MX PA06003677A
Authority
MX
Mexico
Prior art keywords
epo
central
hinge region
mimic
seq
Prior art date
Application number
MXPA/A/2006/003677A
Other languages
Spanish (es)
Inventor
John Ghrayeb
George A Heavner
David M Knight
Bernard J Scallon
Thomas C Nesspor
Chichi Huang
Original Assignee
Centocor Inc
John Ghrayeb
Heavner George
Chichi Huang
David M Knight
Thomas C Nesspor
Scallon Bernard
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, John Ghrayeb, Heavner George, Chichi Huang, David M Knight, Thomas C Nesspor, Scallon Bernard filed Critical Centocor Inc
Publication of MXPA06003677A publication Critical patent/MXPA06003677A/en

Links

Abstract

The present invention relates to at least one novel human EPO mimetic hinge core mimetibody or specified portion or variant, including isolated nucleic acids that encode at least one EPO mimetic hinge core mimetibody or specified portion or variant, EPO mimetic hinge core mimetibody or specified portion or variants, vectors, host cells, transgenic animals or plants, and methods of making and using thereof, including therapeutic compositions, methods and devices.

Description

CENTRAL HUMAN MIMETICUERPOSES OF THE HIPPER REGION, MIMETICS OF ERYTHROPOYETINE, COMPOSITIONS. METHODS AND USES FIELD OF THE INVENTION The present invention relates to central mimetibodies of the hinge region, mammalian EPO mimetics, specific portions and specific variants for biologically active proteins, fragments or ligands, nucleic acids encoding and complementary to a central mimetibody of the hinge region. , EPO mimetic, host cells and methods for making and using same, including, formulations, administration and therapeutic devices.
BACKGROUND OF THE INVENTION Recombinant proteins are an emerging class of therapeutic agents. Such recombinant therapeutic agents have produced advances in the chemical formulation and modification of proteins. Such modifications can potentially improve the therapeutic utility of therapeutic proteins, such as increasing half-lives (for example, by blocking their exposure to proteolytic enzymes), improving biological activity or reducing unwanted side effects. A Such modification is the use of immunoglobulin fragments fused to receptor proteins, such as an enterocept. Therapeutic proteins have also been constructed using the Fc domain to try to provide a longer half-life or to incorporate functions such as Fc receptor binding, protein A binding and complement fixation. A specific and vital role of the mammalian hematopoietic system is the production of erythrocytes or red blood cells, which transport oxygen to the various tissues of the animal's body. The procedure to produce erythrocytes ("erythropoiesis") occurs continuously during the life of the animal to counteract the destruction of erythrocytes. The typical red blood cell has a relatively short life, usually 100 to 120 days. Erythropoiesis is a precisely controlled physiological mechanism by which sufficient numbers of erythrocytes are produced to allow adequate tissue oxygenation, but not so much to impede circulation. It is now known that erythropoiesis is mainly controlled by polypeptide erythropoietin (EPO), an acid glycoprotein. Erythropoietin occurs as the result of the expression of a gene from a single copy located on a chromosome of a mammal. The amino acid sequence for recombinant human EPO ("rHuEPO") is substantially identical to the amino acid sequence for EPO obtained from human urinary sources. However, the glycosylation of rHuEPO differs from that of urinary EPO and human serum EPO.
In a healthy mammal EPO is present in blood plasma at very low concentrations, since the tissues are sufficiently oxygenated by the number of circulating erythrocytes. The present EPO stimulates the production of new erythrocytes to replace those lost due to the aging process. In addition, EPO production is stimulated under hypoxic conditions, where the oxygen supply to body tissues is reduced below normal physiological levels despite adequate perfusion of the tissue by the blood. Hypoxia can be caused by hemorrhage, radiation-induced destruction of erythrocytes, various anemias, high altitude, or long periods of unconsciousness. In contrast, if the number of red blood cells in circulation exceeds that needed for normal tissue oxygenation, the production of EPO is reduced. However, certain disease states involve abnormal erythropoiesis. Recombinant human EPO (rHuEPO) is being used therapeutically in several countries. In the United States, the United States Food and Drug Administration (FDA) has approved the use of rHuEPO to treat anemia associated with end-stage renal disease. Patients who undergo hemodialysis to treat this disorder typically suffer from severe anemia caused by rupture and premature death of erythrocytes as a result of dialysis treatment. EPO is also useful in the treatment of other types of anemia. For example, anemia induced by chemotherapy, anemia associated with myelodysplasia, those associated with several congenital disorders, anemia related to AIDS and anemia associated with prematurity, can be treated with EPO. In addition, EPO may play a role in other areas, such as helping to re-establish a normal hematocrit more quickly in patients with bone marrow transplantation, in patients preparing for autologous blood transfusions, and in patients suffering from overload disorders of iron. Erythropoietin (EPO) is a glycoprotein hormone composed of 165 amino acids and four carbohydrate chains that functions as the main regulator of erythropoiesis, binding to a specific receptor on the surface of erythrocyte precursor cells. This union indicates its proliferation and differentiation into mature red blood cells. The erythropoietin receptor is a 484 amino acid glycoprotein with high affinity for erythropoietin. For the erythropoietin receptor, the homodimerization induced by the ligand may be one of the key events that govern activation. Erythropoietin has a relatively short half-life. Erythropoietin administered intravenously is eliminated at a rate consistent with first-order kinetics with a circulating half-life ranging from approximately 3 to 4 hours in patients with CRF. Within the range of the therapeutic dose, the detectable levels of erythropoietin in the plasma are maintained for at least 24 hours. After subcutaneous administration of erythropoietin, maximum levels in serum they are reached in the course of 5-24 hours and decline slowly after this. The small erythropoietin peptidomimetics were identified by several groups through the screening of random libraries of peptides showing the phage for affinity of the erythropoietin receptor. These sequences have no homology with erythropoietin.
In the functional assays, several of these peptides showed activity, but only 1 / 100,000 of that of the recombinant erythropoietin.
Although several attempts have been made to increase the potency of these peptides by preparing covalent dimers or multimers of the peptidomimetics, these compounds are still 1, 000-10,000 times less active than erythropoietin on a molar basis, and have very short half-lives, which has made them unsuitable for use as therapeutic agents. Accordingly, there is a need to provide improved and / or modified versions of therapeutic EPO proteins, which overcome one or more of these and other problems known in the art.
BRIEF DESCRIPTION OF THE INVENTION The present invention provides central human mimetibodies of the hinge region, EPO mimetics, including modified immunoglobulins, cleavage products and other specified portions and variants thereof, as well as compositions of central mimetibodies of the hinge region, mimetics. of EPO, encoding or complementary nucleic acids, vectors, host cells, compositions, formulations, devices, transgenic animals, transgenic plants and methods for making and using same, as described and / or enabled herein, in combination with which is known in the art. The present invention also provides at least one isolated central mimetibody of the hinge region, EPO mimetic or a specified portion or variant as described herein and / or as is known in the art. The central mimetibody of the EPO mimetic hinge region may optionally comprise at least one CH3 region directly linked to at least one CH2 region directly linked to at least a portion of at least one hinge region or a fragment thereof ( H), directly linked to an optional linker sequence (L), directly linked to at least one EPO (P) mimetic therapeutic peptide, optionally, directly further linked to at least a portion of at least one variable antibody sequence ( V). In a preferred embodiment, a pair of CH3-CH2- hinge-linker-therapeutic peptide region with an N-terminal antibody sequence, the pair is optionally linked by the association or a covalent bond, such as, but not exclusively, at least one Cys-Cys disulfide bond or at least one CH4 or another immunoglobulin sequence. In one embodiment, a central mimetibody of the hinge region, mimic of EPO, comprises formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of an N term of an immunoglobulin variable region, P is at least a bioactive EPO mimic polypeptide, L is at least a linking sequence, H is at least a portion of a variable region of the immunoglobulin, CH2 is at least a portion of a CH2 constant region of the immunoglobulin, CH3 is at least a portion of an immunoglobulin CH3 constant region, m, n, op, q, rys can be, independently, an integer between 0, 1 or 2 and 10, which mimic different types of immunoglobulin molecules, for example, non-exclusively, IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM, IgD, IgE or any subclass thereof and the similar, or any combination thereof. Thus, a central mimetibody of the hinge region, mimetic of the EPO of the present invention, mimics at least a portion of an antibody or an immunoglobulin structure or function, with its inherent properties and functions, while providing a therapeutic peptide and its properties or activities inherent or acquired in vitro, in vivo or in situ. The various portions of the antibody and the therapeutic peptide portions of at least one central mimetic of the hinge region, mimetic of the EPO of the present invention, may vary as described herein in combination with what is known in the art. . The present invention provides, in one aspect, isolated nucleic acid molecules which comprise, are complementary, have a significant identity or hybridize to specific mimetibodies encoding the polynucleotide or specified portions or variants thereof, comprising at least one sequence, domain, portion or specified variant thereof. The present invention further provides recombinant vectors comprising at least one of the nucleic acid molecules of the central isolated mimetibody of the hinge region, mimetic of EPO, host cells containing such nucleic acids and / or recombinant vectors, as well as methods for making and / or using such nucleic acids of the central mimetibody of the hinge region, EPO mimetic, vectors and / or host cells. At least one central mimetibody of the hinge region, mimic of the EPO or a specified portion or variant of the invention, mimics the binding of the P portion of the mimetibody to at least one ligand, or has at least one biological activity of at least one a protein, subunit, fragment, portion or any combination thereof.
The present invention also provides at least one central isolated mimetibody of the hinge region, mimetic of EPO or a portion or variant specified as described herein and / or as is known in the art, wherein the central mimetic of the hinge region, mimic of the EPO or a specified portion or variant has at least one activity, such as, but not exclusively, the known biological activities of at least one bioactive peptide or polypeptide corresponding to the P portion of Formula I Therefore, a central mimetic body of the hinge region, EPO mimetic can be screened for the corresponding activity according to known methods, such as at least one neutralizing activity towards a protein or fragment thereof. The present invention also provides at least one composition comprising (a) at least one central isolated mimetibody of the hinge region, EPO mimic or a specified portion or variant encoding a nucleic acid and / or a central mimetic of the region of hinge, mimic of EPO 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 may optionally further comprise at least one compound, protein or additional composition. The present invention also provides at least one method for expressing at least one central mimetibody of the hinge region, EPO mimic or a specified portion or variant in a host cell, comprising culturing a host cell as described herein and / or as is known in the art, under conditions wherein at least one central mimetic of the host region hinge, mimetic of the EPO or a specified portion or variant, is expressed in detectable and / or recoverable amounts. The present invention further provides at least one central mimetic of the hinge region, mimetic of EPO, a specified portion or variant in a method or composition, when administered in a therapeutically effective amount for modulation, to treat or reduce the symptoms of at least one bone and joint disorder, a cardiovascular disorder, a dental or oral disorder, a dermatological disorder, an ear, nose or throat disorder, an endocrine or metabolic disorder, a gastrointestinal disorder, a gynecological disorder, a disorder hepatic or biliary, an obstetric disorder, a hematological disorder, an immunological or allergic disorder, an infectious disease, a musculoskeletal disorder, an oncological disorder, a neurological disorder, a nutritional disorder, an ophthalmological disorder, a pediatric disorder, a poisoning disorder , a psychiatric disorder, a kidney disorder, an after pulmonary lathe or any other known disorder. (See, for example, The Merck Manual, 17th ed., Merck Research Laboratories, Merck and Co., Whitehouse Station, NJ (1999), fully incorporated herein by reference), as needed in many different conditions, such as as non-exclusive, before, after, or during a related disease or treatment condition, as is known in the art. The present invention further provides at least one central mimetic of the hinge region, EPO mimic, a portion or variant specified in a method or composition, when administered in a therapeutically effective amount, for modulation, to treat or reduce the symptoms of at least one immune, cardiovascular, infectious, malignant and / or neurological disease in a cell, tissue, organ, animal or patient and / or, as needed in many different conditions, such as, in a non-exclusive manner, prior to , subsequent to, or during a related disease or treatment condition, as is known in the art and / or as described herein. The present invention also provides at least one composition, device and / or method for delivering a therapeutically or prophylactically effective amount of at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant, in accordance with present invention. The present invention further provides at least one anti-idiotype antibody to at least one central mimetic of the hinge, mimetic region of the EPO of the present invention. The anti-idiotype antibody includes any protein or peptide that contains a molecule that comprises at least a portion of an immunoglobulin molecule, such as, non-exclusively, at least one complementary determining region (CDR) of a heavy or light chain or a ligand-binding portion thereof, a variable region of heavy chain or light chain, a constant chain region heavy or light chain, a region of the framework or any portion thereof, which binds competitively to a region that binds to the EPO receptor of at least one central mimetibody of the hinge region, mimic of the EPO of the present invention. Such idiotype antibodies of the invention may 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 also provides at least one isolated nucleic acid molecule comprising, complementing or hybridizing to a polynucleotide encoding at least one anti-idiotype antibody of a central mimetic of the hinge region, EPO mimic, comprising at least one sequence, domain, portion or specified variant thereof. The present invention further provides recombinant vectors comprising the nucleic acid molecules encoding the anti-idiotype antibody of the central mimetibody of the hinge region, mimic of EPO, host cells containing such nucleic acids and / or recombinant vectors, as well as methods for making and / or using such nucleic acids of the anti-idiotype antibody, vectors and / or host cells. The present invention also provides at least one method for expressing at least one central mimetibody of the hinge region, mimetic of EPO, or an anti-idiotype antibody of the central mimetibody of the hinge region, mimic of EPO, in a host cell, comprising culturing a host cell as described herein, under conditions wherein at least one central mimetibody of the hinge region, mimetic of EPO or an anti-idiotype antibody is expressed in detectable and / or recoverable amounts. The present invention also provides at least one composition comprising (a) a nucleic acid encoding an isolated central mimetibody of the hinge region, mimic of EPO and / or a central mimetibody of the hinge region, mimic of EPO, as described herein; and (b) a suitable carrier or diluent. The carrier or diluent can optionally be pharmaceutically acceptable, according to the known carriers or diluents. The composition may optionally further comprise at least one compound, protein or additional composition. The present invention further provides at least one method or composition of a central mimetic of the hinge region, mimic of EPO, for administering a therapeutically effective amount to modulate or treat at least one condition related to the protein in a cell, tissue, organ, animal or patient and / or, before, of, subsequent to, or during a related condition, as is known in the art and / or as described herein.
The present invention also provides at least one composition, device and / or method for delivering a therapeutically or prophylactically effective amount of at least one central mimetibody of the hinge region, EPO mimetic, according to the present invention. The present invention further provides at least one method or composition of a central mimetibody of the hinge region, EPO mimetic, for diagnosing at least one condition related to EPO in a cell, tissue, organ, animal or patient and / or , before, after, or during a related condition, as is known in the art and / or as described herein. The present invention also provides at least one composition, device and / or method for providing the diagnosis of at least one central mimetibody of the hinge region, EPO mimetic, according to the present invention. In one aspect, the present invention provides at least one centrally isolated human mimetibody of the hinge region, EPO mimetic, comprising at least one P (n) region, comprising at least a portion of at least one of the SEQ ID NOS: 1-30, for example, as presented in Table 1 below, or optionally with one or more substitutions, deletions or insertions as described herein or as is known in the art. In another aspect, the present invention provides at least one centrally isolated human mimetibody of the hinge region, EPO mimetic, wherein the central mimetibody of the region of hinge, EPO mimetic specifically binds to at least one epitope comprising at least 1-3 of at least one ligand or binding region, ligand which binds to at least a portion of at least one of SEQ ID NOS : 1-30 as presented in Table 1 below, or optionally with one or more substitutions, deletions or insertions, as described herein or as is known in the art. The at least one central mimetibody of the hinge region, EPO mimetic, can optionally further perform at least one of: binding to the protein with an affinity of at least one selected of at least 10"9 M, at least 10"10 M, at least 10" 11 M or at least 10"12 M; substantially neutralize at least one activity of at least one protein or portion thereof. Also provided is an isolated nucleic acid encoding at least one centrally isolated human mimetibody of the hinge region, EPO mimetic; 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-1, COS-7, HEK293, BHK21, CHO, BSC-1, Hep G2, 653, SP2 / 0, 293, HeLa, myeloma or lymphoma cells, or any derived cells, immortalized or transformed from them. A method is also provided for producing at least one central mimetibody of the hinge region, EPO mimetic, which comprises translating the nucleic acid encoding the central mimetibody of the hinge region, low EPO mimetic. conditions in vitro, in vivo or in situ, so that the central mimetibody of the hinge region, EPO mimic, is expressed in detectable or recoverable amounts. Also provided is a composition comprising at least one centrally isolated human mimetibody of the hinge region, EPO mimetic and at least one pharmaceutically acceptable carrier or diluent. The composition may 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 drug for the cardiovascular system (CV), a drug for the central nervous system. (CNS), a drug for autonomic nervous system (ANS), a drug for the respiratory tract, a drug to the gastrointestinal (Gl), a hormonal drug, a drug for fluid balance or electrolyte, a hematologic drug , an anticancer drug, a drug for immunomodulation, nasal ophthalmic drug, otic or a topical drug, a nutritional drug, an antagonist drug TNF, antirheumatic, a muscle relaxant, narcotic, nonsteroidal anti-inflammatory (nthe), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial, an antisoriático, a corticosteroid, a eroide anabolic, an erythropoietin, an immunization, an immunoglobulin, an immunosuppressive, a growth hormone, a drug for hormone replacement, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, a medication for asthma, an agonist beta, an inhaled steroid, an epinephrine or analogue, a cytokine or a cytokine antagonist. The present invention further provides an antibody or anti-idiotype fragment that specifically binds to at least one central mimetibody of the hinge region, mimetic of the EPO of the present invention. A method is also provided 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 mimetibody central to the hinge region, mimic of the EPO of the invention with the cell, tissue, organ or animal. The method may further optionally comprise using an effective amount of 0.001-50 mg / kilogram of the cells, tissue, organ or animal. The method may further comprise optionally use the contact or administration by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intraarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebelar, intracerebroventricular, intracolic, intracervical, intragastric , ntrahepático, intramyocardial, intraósteo, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, ntrarretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal or transdermal. The method may further optionally comprise administering before, concurrently or after (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 brand or reporter. , an anti-infective drug, a drug for the cardiovascular system (CV), a drug for the central nervous system (CNS), a drug for the autonomic nervous system (ANS), a drug for the respiratory tract, a drug for the gastrointestinal tract (Gl), a hormonal drug, a drug for fluid balance or electrolyte, a hematologic drug, an antineoplastic drug, a drug for immunomodulation, nasal ophthalmic drug, otic or a topical drug, a nutritional drug, a TNF antagonist drug, antirheumatic, muscle relaxant, narcotic, nonsteroidal anti-inflammatory (NSAID), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial, an antisoriático, a corticosteroid, an anabolic steroid, an erythropoietin, an immunization , an immunoglobulin, an immunosuppressant, a growth hormone, a hormone replacement drug, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, a medication for asthma, a beta agonist, an inhaled steroid, an epinephrine or the like, a cytokine or a cytokine antagonist. Also provided is a medical device, comprising at least one isolated central human mimetibody of the hinge region, mimetic of the EPO of the invention, wherein the device is suitable for contacting or administering the at least one central mimetibody of the hinge, EPO mimetic region by at least one selected parenteral, subcutaneous, intramuscular mode, intravenous, intraarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavity, intracellular, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intrahepatic, intramyocardial, intradose, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal , intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal or transdermal. Also provided is an article of manufacture for pharmaceutical or diagnostic use in humans, comprising a packaging material and a container comprising a solution or a lyophilized form of at least one isolated human mimetibody central to the hinge region, mimetic of the EPO of the present invention. The article of manufacture may optionally comprise having the container as a component of a parenteral, subcutaneous, intramuscular, intravenous, intraarticular, intrabronchial, intraabdominal, intracapsular, intracartilage, intracavity, intracelial, intracelebelar, intracerebroventricular, intracolic, intracervical, delivery or delivery system or device. intragastric, intrahepatic, intramyocardial, intratracheal, intrapelvic, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, Intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal or transdermal. A method is also provided for producing at least one centrally isolated human mimetibody of the hinge region, mimic of the EPO of the present invention, which comprises providing a host cell or a transgenic animal or a transgenic plant or a plant cell capable of express in recoverable quantities, the central mimetibody of the hinge region, EPO mimic. In the present invention, there is further provided at least one central mimetibody of the hinge region, mimic of the EPO produced by the above method. The present invention also provides at least one method for expressing at least one central mimetibody of the hinge region, EPO mimic, or an anti-idiotype antibody, in a host cell, which comprises culturing a host cell as described herein under conditions wherein at least one central mimetibody of the hinge region, EPO mimic is expressed in detectable and / or recoverable amounts. The present invention further provides any invention described herein.
DETAILED DESCRIPTION OF THE INVENTION The present invention provides mimetibodies or recombinant and / or synthetic specified portions or variants, as well as coding nucleic acid compositions and molecules comprising at least one polynucleotide encoding at least one central mimetic of the hinge region, mimic of EPO. Such mimetibodies or specified portions or variants of the present invention comprise specific sequences of central mimetibodies of the hinge region, EPO mimetics, domains, fragments and variants thereof, and methods for making and using nucleic acids and mimetibodies. or specified portions or variants, including compositions, methods and therapeutic devices. The present invention also provides at least one isolated central mimetibody of the hinge region, EPO mimetic or a specified portion or variant as described herein and / or as is known in the art. The central mimetibody of the hinge region, EPO mimetic may optionally comprise at least one CH3 region directly linked to at least one CH2 region directly linked to at least one hinge region or fragment thereof (H), directly linked to an optional linker sequence (L), directly linked to at least one therapeutic peptide (P), linked directly in addition optionally with at least a portion of at least one variable sequence of antibody (V). In a preferred embodiment, a central mimetibody of the hinge region, mimic of EPO, comprises formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of a N-terminus of a immunoglobulin variable region, P is at least one bioactive peptide, L is a polypeptide that provides structural flexibility, allowing the mimetibody to have alternating binding orientations and properties, H is at least a portion of a variable immunoglobulin 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 and s can independently be an integer between 0, 1 or 2 and 10, which mimic different types of immunoglobulin molecules, for example, non-exclusively, IgG1, IgG2, IgG3, IgG4, IgA, IgM, IgD, IgE and the like, or combinations thereof. where m = 1, can be linked to other monomers by covalent bonding or association, such as, but not limited to, a disulfide bond Cys-Cys or another immunoglobulin sequence. The central mimetibody of the hinge, mimetic region of the EPO of the present invention mimics an antibody structure with its inherent properties and functions, while providing a therapeutic peptide and its properties or activities inherent or acquired in vitro, live or in situ. The various portions of the antibody and the therapeutic peptide portions of at least one central mimetibody of the hinge region, mimetic of the EPO of the present invention, may vary as described herein in combination with what is known in the art. As used herein, a "central mimetibody of the hinge region, EPO mimetic," "a central mimetibody portion of the hinge region, EPO mimetic," or "central mimetic antibody fragment of the region." of hinge, EPO mimic "and / or" variant of the central mimetibody of the hinge region, mimic of EPO "and the like, mimics, has or simulates at least one ligand binding or at least one biological activity of at least a protein, such as binding or activity of the ligand in vitro, in situ and / or preferably in vivo, such as, but not limited to, at least one of SEQ ID NOS: 1-30. For example, a central mimetibody of the hinge region, suitable EPO mimic, a specified portion or variant of the present invention, can be attached to at least one protein ligand and includes at least one protein ligand, a receptor, receptor soluble and similar. A central mimetibody of the hinge region, mimetic of the appropriate EPO, specified portion or variant can also modulate, increase, modify, activate at least one protein receptor or signaling or other measurable or detectable activity. The mimetibodies useful in the methods and compositions of the present invention are characterized by a suitable binding affinity. to the ligands or receptors of the protein and optionally and preferably have low toxicity. In particular, a central mimetibody of the hinge region, EPO mimetic, wherein the individual components, such as the portion of the variable region, the constant region (without a CH1 portion) and the framework, or any portion of the (for example, a portion of the J, D or V regions of the heavy or light variable chain, at least a portion of at least one hinge region, the constant heavy or light chain chain and the like), so Individual and / or collective, optionally and preferably possesses 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 relief of symptoms from good to excellent and low toxicity. Low immunogenicity and / or high affinity, as well as other undefined properties, can contribute to the therapeutic results achieved. "Low immunogenicity" is defined herein as elevating significant HAMA, HACA or HAHA responses by less than about 75%, or preferably by 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 treated patients and / or raising the low titres in the treated patient (less than about 300, preferably less than about 100 , measured with an enzyme immunoassay with double antigen) (see, for example, Elliott et al., Lancef 344: 1125-1127 (1994)).
Utility The isolated nucleic acids of the present invention can be used for the production of at least one central mimetic of the hinge region, EPO mimetic, fragment or specified variant thereof, which can be used to effect in a cell, tissue, organ or animal (including mammals and humans), to modulate, treat, alleviate, help prevent the occurrence of or reduce the symptoms of, at least one condition related to a protein, selected, non-exclusively, from at least one of an immune disorder or disease, cardiovascular disorder or disease, an infectious, malignant and / or neurological disorder or disease, an anemia; an immune / autoimmune condition; and / or carcinogenic / infectious, as well as other known or specified protein-related conditions. Such a method may comprise administering an effective amount of a composition or a pharmaceutical composition comprising at least one central mimetic of the hinge region, EPO mimetic or a specified portion or variant to a cell, tissue, organ, animal or patient in need for such modulation, treatment, relief, prevention or reduction in symptoms, effects or mechanisms. The effective amount may comprise an amount of about 0.0001 to 500 mg / kg per single or multiple administration, or to achieve a serum concentration of 0.0001-5000 μg / ml of serum concentration per single or multiple administration, or any range or value effective in it, how it is done and determined using known methods, as described herein or known in the relevant arts.
Appointments All publications and patents cited herein are incorporated herein by reference in their entirety, since they show the state of the art at the time of the present invention and / or provide a description and enablement of the present invention. The publications refer to any scientific or patent publication, or any other information available in any media format, including all registered, electronic or printed formats. The following references are hereby incorporated by reference in their entirety: Ausubel, et al., Ed., Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY, NY (1987-2003); Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor, NY (1989); Harlow and Lane, Antibodies, a Laboratory Manual, Cold Spring Harbor, NY (1989); Colligan, et al., Eds., Current Protocols in Immunology, John Wiley & Sons, Inc., NY (1994-2003); Colligan et al., Current Protocols in Protein Science, John Wiley & Sons, NY, NY, (1997-2003).
Mimetibodies of the present invention The central mimetibody of the hinge, mimetic region of EPO may optionally comprise at least one linked CH3 region directly with at least one CH2 region directly linked to at least a portion of at least one fragment of the hinge region (H), such that it comprises at least one region of central hinge, directly linked to an optional linker sequence (L) , directly linked to at least one therapeutic peptide (P), optionally further linked, directly to at least a portion of at least one variable antibody sequence (V). In a preferred embodiment, a pair of CH3-CH2-H-L-V, the pair linked by association or the covalent bond. Thus, a central mimetibody of the hinge region, mimetic of the EPO of the present invention, mites an antibody structure with its inherent properties and functions, while providing a therapeutic peptide and its inherent or acquired properties or activities in vitro, n live or in situ. The various portions of the antibody and the therapeutic peptide portions of at least one central mimetibody of the hinge region, mimetic of the EPO of the present invention, may vary as described herein in combination with what is known in the art. The mimetibodies of the present invention thus provide at least one suitable property compared to known proteins, such as, non-exclusively, at least one of increased half-life, increased activity, more specific activity, increased strength, increased or decreased inactivation rate, a selected activity or a more suitable subset of activities, less immunogenicity, quality or increased duration of at least one desired therapeutic effect, less side effects and the like. Fragments of the mimetibodies according to Formula (I), can be produced by enzymatic cleavage, synthetic or recombinant techniques, as is known in the art and / or as described herein. The 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 the mimetibodies can be chemically linked by conventional techniques, or they can be prepared as a contiguous protein using genetic engineering techniques. For example, a nucleic acid encoding at least one of the constant regions of a human antibody chain can be expressed to produce a contiguous chain for use in the mimetibodies of the present invention. See, for example, Ladner et al., U.S. Patent. No. 4,946,778 and Bird, R. E. et al., Science, 242: 423-426 (1988), with respect to single chain antibodies. As used herein, the term "human mimetibody" refers to an antibody in which substantially every part of the protein is expected (eg, EPO mimetic peptide, framework, CL, CH domains (e.g. , CH2, CH3), hinge (VL, VH)), is substantially non-immunogenic in humans, with only minor changes or variations in the sequence. Such changes or variations optionally, and preferably, retain or reduce immunogenicity in humans relative to unmodified human antibodies, or mimetibodies of the present invention. Thus, a human antibody and the central mimetibody of the hinge region, mimetic of the corresponding EPO of the present invention, is distinct from a chimeric or humanized antibody. It is noted that a human antibody and a central mimetibody of the hinge region, mimic of EPO, can be produced by an animal or non-human cell that is capable of expressing the genes of human immunoglobulins (e.g., heavy chain and / or chain). light). Human mimetibodies that are specific for at least one ligand or protein receptor thereof, can be designed against an appropriate ligand, such as a receptor or ligand isolated from the protein and / or EPO or a portion thereof (including molecules synthetic, such as synthetic peptides). The preparation of such mimetibodies is performed using known techniques to identify and characterize the ligand binding regions or sequences of at least one protein or portion thereof. In a preferred embodiment, at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention, is produced by at least one cell line, a mixed cell line, an immortalized cell or a clonal population of immortalized and / or cultured cells. The immortalized cells that produce a protein, can be produced using appropriate methods. Preferably, the at least one central mimetic of the hinge region, mimic of the EPO or a specified portion or variant, is generated by providing nucleic acids or vectors comprising derived DNA having a sequence substantially similar to at least one site of the invention. human immunoglobulin which is functionally rearranged, or which can undergo a functional rearrangement, and which further comprises a mimetibody structure as described herein, for example, non-exclusively, Formula (I), wherein the portions of the variable regions C and N terminals can be used for V, the hinge regions for H, CH2 for CH2 and CH3 for CH3, as is known in the art. The term "functionally rearranged", as used herein, refers to a segment of a nucleic acid from an immunoglobulin site that has undergone V (D) J recombination., thereby producing an immunoglobulin gene encoding an immunoglobulin chain (e.g., heavy chain), or any portion thereof. A functionally rearranged immunoglobulin gene can be identified directly or indirectly using suitable methods, such as, for example, nucleotide sequencing, hybridization (e.g., Southern blotting, Northern blotting), using probes that can anneal to the coding junctions between the segments of the gene or enzymatic amplification of immunoglobulin genes (eg, polymerase chain reaction) with primers that can anneal to the coding junctions between the segments of the gene. If a cell produces a central mimetibody of the hinge region, EPO mimic or a portion or variant comprising a particular variable region or a variable region comprising a particular sequence (eg, at least one P sequence), it can also Determine using appropriate methods. The mimetibodies, portions and variants specified in the present invention, can also be prepared using at least one nucleic acid encoding a central mimetibody of the hinge region, EPO mimic or a specified portion or variant, 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 the sequences encoding the antibody. See, for example, non-exclusively, US Patents. Nos. 5,827,690; 5,849,992; 4,873,316; 5,849,992; 5,994,616; 5,565,362; 5,304,489, and the like, each of which is fully incorporated herein by reference. The mimetibodies, portions and variants specified herein, can be further prepared using at least one nucleic acid encoding a central mimetibody of the hinge region, mimetic of EPO or a specified portion or variant, to provide transgenic plants and cells of cultivated plants (for example, non-exclusively, tobacco and corn) that produce such mimetibodies, portions or variants specified in the parts of the plant or in the cultivated cells thereof. As a non-limiting example, transgenic tobacco leaves expressing recombinant proteins have been used successfully to provide large amounts of recombinant proteins, for example, using an inducible promoter. See, for example, Cramer et al., Curr. Top. Microbol. Immunol. 240: 95-118 (1999) and the references cited therein. Also, transgenic corn has been used to express mammalian proteins at levels of commercial production, with biological activities equivalent to those produced in other recombinant systems or purified from natural sources. See, for example, Hood et al., Adv. Exp. Med. Biol. 464: 127-147 (1999) and the references cited therein. Antibodies in large quantities have also been produced from seeds of transgenic plants, including antibody fragments, such as single chain mimetibodies (scFv), including tobacco seeds and potato tubers. See, for example, Conrad et al., Plant Mol. Biol. 38: 101-109 (1998) and the references cited therein. Thus, the mimetibodies, portions and variants specified of the present invention, can also be produced using transgenic plants, according to known methods. See also, for example, Fischer et al., Biotechnol. Appl. Biochem. 30: 99-108 (October, 1999), Ma et al., Trends Biotechnol. 13: 522-7 (1995); Ma et al., Plant Physio. 109: 341-6 (1995); Whitelam et al., Biochem. Soc. Trans. 22: 940-944 (1994); and the references cited in the same. The above references are incorporated herein by reference in their entirety. The mimetibodies of the invention can bind ligands of human protein with a wide variety of affinities (KD). In a preferred embodiment, at least one central human mimetibody of the hinge region, mimetic of the EPO of the present invention can optionally be linked to at least one protein ligand with high affinity. For example, at least one central mimetic of the hinge, mimetic region of the EPO of the present invention can be linked to at least one protein ligand with a KD equal to or less than about 10 ~ 7 M or, more preferably, with a KD equal to or less than approximately 0.1-9.9 (or any interval or value therein) X 10"7, 10 ~ 8, 10"9, 10" 10, 10"11, 10" 12 or 10"13 M, or any interval or value therein The affinity or strength of a central mimetibody of the hinge region, mimic EPO for at least one protein ligand can be determined experimentally using any suitable method, for example, as used to determine the affinity or binding strength of the antibody-antigen (See, for example, Berzofsky, et al., "Antibody-Antigen Interactions", In Fundamental Immunology, Paul, WE, Ed., Raven Press: New York, NY (1984); Kuby, Janis Immunology, W. H. Freeman and Company: New York, NY (1992); and methods described therein). The measured affinity of a particular central mimetibody interaction of the hinge region, mimic of the EPO-ligand, may vary if measured under different conditions (for example, salt concentration, pH). Thus, affinity measurements and other ligand binding parameters (e.g., KD, Ka, K), are preferably made with standardized solutions of the central mimetibody of the hinge region, mimic of EPO and the ligand, and a standardized shock absorber, such as the shock absorber described herein.
Nucleic Acid Molecules Using the information provided herein, such as nucleotide sequences encoding at least 90-100% of the contiguous amino acids of at least one of SEQ ID NOS: 1-30, as well as at least a portion of an antibody, wherein the above sequences are inserted as the sequence P of Formula (I) to provide a central mimetic of the hinge region, mimetic of the EPO of the present invention, which further comprises fragments, specified variants or sequences of consensus thereof, or a deposited vector comprising at least one of these sequences, a nucleic acid molecule of the present invention encoding at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant, it can be obtained using the methods described herein or as are known in the art. The nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, ARNhn, tRNA or any another form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or synthetically produced, or any combination thereof. The DNA can be triple-stranded, double-stranded or single-stranded, or any combination thereof. A portion of at least one strand of DNA or RNA can be a coding strand, also known as the sense strand or it can be the non-coding strand, also referred to as the antisense strand. The isolated nucleic acid molecules of the present invention may 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 central mimetic of the hinge region, mimic of EPO or a specified portion or variant; and nucleic acid molecules comprising a nucleotide sequence substantially different from those described above, but which, due to the degeneracy of the genetic code, still encode at least one central mimetibody of the hinge region, EPO mimetic as described in US Pat. present and / or as is known in the art. Of course, the genetic code is well known in the art. Thus, it would be routine for someone skilled in the art to generate such degenerate variants of nucleic acids encoding the central specific mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention. See, for example, Ausubel, et al., supra, and such nucleic acid variants are included in the present invention. As indicated herein, the nucleic acid molecules of the present invention comprising a nucleic acid encoding a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant may include, but are not limited to, , those encoding the amino acid sequence of a central mimetibody fragment of the hinge region, EPO mimetic, by itself; the coding sequence for all the central mimetibody of the hinge region, mimic of the EPO or a portion thereof; the coding sequence for a central mimetibody of the hinge region, mimic of EPO, fragment or portion, as well as additional sequences, such as the coding sequence for at least one signal or fusion leader peptide, with or without the sequences additional encoders mentioned above, such as an intron, together with additional non-coding sequences, including, but not limited to, the non-coding 5 'and 3' sequences, such as transcribed, untranslated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals (e.g., ribosome binding and mRNA stability); and additional coding sequences that encode additional amino acids, such as those that provide additional functionalities. Thus, the sequence encoding a central mimetibody of the hinge region, EPO mimic or a specified portion or variant, can be fused to a marker sequence, such as a peptide-encoding sequence that facilitates the purification of the central fused mimetibody of the hinge region, EPO mimetic or a specified portion or variant, comprising a fragment or moiety of the core mimetic the hinge region, EPO mimic.
Polynucleotides that selectively hybridize to a polynucleotide as described herein. The present invention provides isolated nucleic acids that hybridize under conditions of selective hybridization to a polynucleotide described herein, or others described herein, including variants or specified portions of the same. Thus, the polynucleotides of this embodiment can be used to isolate, detect and / or quantify the nucleic acids comprising such polynucleotides. Hybridization conditions of low or moderate stringency are typically, but not exclusively, employed with sequences that have a reduced sequence identity relative to the complementary sequences. The conditions of moderate and high rigor can be used optionally for sequences of greater identity. Low stringency conditions allow for selective hybridization of sequences that have approximately 40-99% sequence identity and can be used to identify orthologous or paralogical sequences.
Optionally, the polynucleotides of this invention will encode at least a portion of a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant encoded by the polynucleotides described herein. The polynucleotides of this invention encompass nucleic acid sequences that can be employed for selective hybridization to a polynucleotide encoding a central mimetic of the hinge region, EPO mimetic or a specified portion or variant of the present invention. See, for example, Ausubel, supra; Colligan, supra, each one fully incorporated in the present as 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 is well known in the art. The nucleic acids may conveniently comprise sequences in addition to a polynucleotide of the present invention. For example, a multiple cloning site comprising one or more restriction sites of the endonuclease can be inserted into the nucleic acid to aid in the isolation of the polynucleotide. Also, translatable sequences can be inserted to assist in the isolation of the translated polynucleotide of the present invention. For example, a sequence marker with hexahistidine, 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 a linker for cloning and / or for the expression of a polynucleotide of the present invention. Additional sequences may be added to such cloning and / or expression sequences to optimize their function in cloning and / or expression, to aid in the isolation of the polynucleotide or to improve the introduction of the polynucleotide into a cell. The use of cloning vectors, expression vectors, adapters and linkers is well known in the art. See, for example, Ausubel, supra; or Sambrook, supra.
Recombinant methods for constructing nucleic acids Nucleic acid compositions isolated from this The invention, such as RNA, cDNA, genomic DNA or any combination thereof, can be obtained from biological sources using any of several cloning methodologies known to those skilled in the art. In some embodiments, oligonucleotide probes that hybridize selectively, under conditions of adequate stringency to the polynucleotides of the present invention, are used to identify the desired sequence in a cDNA or genomic DNA library. Isolation of RNA and construction of cDNA libraries and genomic, is well known to those with ordinary experience in the art. (See, for example, Ausubel, supra; or Sambrook, supra).
Synthetic Methods for Building Nucleic Acids The isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by known methods (see, for example, Ausubel, et al., Supra). Chemical synthesis generally produces a single-stranded oligonucleotide, which can be converted to double-stranded DNA by hybridization with a complementary sequence, by polymerization with a DNA polymerase using the single strand as a template. One skilled in the art will recognize that although chemical synthesis of DNA can be limited to sequences of approximately 100 or more bases, longer sequences can be obtained by linking shorter sequences.
Recombinant expression cassettes The present invention also 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 genomic sequence, which encodes a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention, may be used for build a cassette of recombinant expression that can be introduced into at least one cell desired host. A recombinant expression cassette will typically comprise a polynucleotide of the present invention operably linked to transcriptional initiation regulatory sequences that will direct transcription of the polynucleotide in the intended host cell. Heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct the expression of the nucleic acids of the present invention. In some embodiments, the isolated nucleic acids serving as a promoter, enhancer or other elements can be introduced in the appropriate position (upstream, downstream or in an intron) of a non-heterologous form of a polynucleotide of the present invention, to overregulate or deregulate the expression of a polynucleotide of the present invention. For example, endogenous promoters can be altered in vivo or in vitro by mutation, deletion and / or substitution, as is known in the art. A polynucleotide of the present invention can be expressed in any sense or antisense orientation as desired. It will be appreciated that control of gene expression in the sense or antisense orientation can have a direct impact on the observable characteristics. Another method of suppression is sense suppression. The introduction of a nucleic acid configured in the sense orientation, has not been shown to be an effective means by which to block the transcription of the target genes.
Vectors and Host Cells The present invention also relates to vectors that include isolated nucleic acid molecules of the present invention, host cells that are genetically engineered with recombinant vectors, and the production of at least one central mimetic of the hinge region. , mimetic of EPO or a portion or variant specified by recombinant techniques, as is well known in the art. See, for example, Sambrook, et al., Supra; Ausubel, et al., Supra, each fully incorporated herein by reference. The polynucleotides can optionally be linked to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced into the 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 the host cells. The DNA insert must be operatively linked to an appropriate promoter. The expression constructs will additionally contain sites optionally for at least one of the start of transcription, 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 starting at the beginning and a terminating Colon (eg, UAA, UGA or UAG) appropriately placed at the end of the mRNA to be translated, with preferred UAA and UAG for cell expression of mammal or eukaryotes. The expression vectors, preferably, but optionally, will include at least one selectable marker. Such labels include, for example, non-exclusively, resistance to methotrexate (MTX), dihydrofolate reductase (DHFR, U.S. Patent 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, U.S. Patent Nos. 5,122,464; 5,770,359; 5,827,739), for the culture of eukaryotic cells and genes for resistance to tetracycline or ampicillin for cultures in E. coli and other bacteria or prokaryotes (the above patents are fully incorporated herein by reference.) The culture media and appropriate conditions for the host cells described above are well known in the art, and suitable vectors will be readily apparent to the skilled artisan. a host cell can be made by transfection with calcium phosphate, transfection mediated by DEAE-dextran, transfection dyed by a cationic lipid, 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 central mimetibody of the hinge region, mimetic of the EPO or a specified portion or variant of the present invention, may be expressed in a modified form, such as a fusion protein, and may include not only secretory signals, but also additional heterologous functional regions. For example, a region of additional amino acids, particularly charged amino acids, may be added to the N terminus of a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant, to improve stability and persistence in the host cell, during purification or during subsequent handling and storage. Also, peptide portions can be added to a central mimetibody of the hinge region, mimetic of the EPO or a specified portion or variant of the present invention to facilitate purification. Such regions can be removed prior to the final preparation of a central mimetibody of the hinge region, EPO mimetic 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 aware of numerous expression systems available for the expression of a nucleic acid encoding a protein of the present invention. Illustrative cell cultures useful for the production of the mimetibodies, specified portions or variants thereof, are the mammalian cells. Mammalian cell systems are often in the form of monolayers of cells, although suspensions or bi-reactors of mammalian cells can also be used. Several suitable host cell lines capable of expressing intact glycosylated proteins have been developed in the art, and include cell lines COS-1 (eg, ATCC CRL 1650), COS-7 (eg, ATCC CRL-1651), HEK293, BHK21 (e.g., ATCC CRL-10), CHO (e.g., ATCC CRL 1610, DG-44) and BSC-1 (e.g., ATCC CRL-26), hepG2 cells, P3X63Ag8.653 cells, SP2 / 0- Ag14, 293, HeLa cells and the like, which are readily available from, for example, the 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 (Accession number of ATCC CRL-1580) and SP2 / 0-Ag14 cells (Accession number of ATCC 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., SV40 late or early promoters, the CMV promoter (e.g., U.S. Patent Nos. 5,168,062; 5,385,839), a tk HSV promoter, a pgk (phosphoglycerate kinase) promoter, an EF-1 alpha promoter ( for example, U.S. Patent No. 5,266,491), 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., a large poly A T Ag addition site of SV40), and transcription termination sequences. See, for example, Ausubel et al., Supra; Sambrook, et al., Supra. Other cells useful for the production of the nucleic acids or proteins of the present invention are known and / or available, for example, from the Catalog of Cell Lines and Hybridomas of the American Type Culture Collection (www.atcc.org) or others well-known or commercial sources. When eukaryotic host cells are employed, the polyadenylation termination or transcription sequences are typically incorporated into the vector. An example of a terminator sequence is the polyadenylation sequence of the bovine growth hormone gene. The sequences for the exact splicing of the transcript can also be included. An example of a splicing sequence is the VP1 intron SV40 (Sprague, et al., J. Virol. 45: 773-781 (1983)). Additionally, gene sequences for controlling replication in the host cell, as is known in the art, can be incorporated into the vector.
Purification of a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant thereof A central mimetibody of the hinge region, mimetic of the EPO or a 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 chromatography with lectin. High performance liquid chromatography ("HPLC") can also be used for purification. See, e.g., Colligan, Current Protocols in Immunology, or Current Protocols in Protein Science, John Wiley & Sons, NY, NY, (1997-2003), for example, Chapters 1, 4, 6, 8, 9, 10, each fully incorporated herein by reference. The mimetibodies or the specified portions or variants of the present invention include naturally purified products, synthetic chemical process products and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast cells, from higher plants, of insect and mammals. Depending on the host employed in a recombinant production process, the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention, may be glycosylated or non-glycosylated, with glycosylated being 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 fully incorporated herein by reference.
Mimetibodies, fragments v / or variants specified The isolated mimetibodies of the present invention comprise a central mimetibody of the hinge region, mimic of the EPO or a specified portion or variant encoded by any of the polynucleotides of the present invention, as discussed with more detail herein, or any central mimetibody of the hinge region, mimetic of EPO isolated or prepared or a specified portion or variant thereof. Preferably, the central mimetic of the hinge region, mimetic of the EPO or the portion or variant that binds to the ligand, binds to at least one ligand or receptor of the EPO protein and, therefore, provides at least one biological activity of the EPO of the corresponding protein or a fragment thereof. Different therapeutic or diagnostically significant proteins, are well known in the art and suitable assays or biological activities of such proteins are well known in the art. Non-limiting examples of the EPO mimetic peptides suitable for this invention appear in Table 1 below. These peptides can be prepared by the methods described and / or known in the art. A single-letter amino acid abbreviations are used in the most of the cases. The X in these sequences (and through this specification, unless otherwise specified in a particular case), means that any of the 20 natural or known amino acid residues or known derivatives thereof may be present, or any known modified amino acid thereof. Any of these peptides may be linked in series (ie, sequentially), with or without linkers, and a few examples linked in series are provided in the Table. The binders are listed as "?", And can be any of the binders described herein. The series repeats and the binders are shown separately by dotted lines for clarity. Any peptide that contains a cysteinyl residue can be optionally cross-linked with another Cys-containing peptide, either or both of which can be linked to a carrier. A few cross-linked examples are provided in the Table. Any peptide having more than one Cys residue can form an intrapeptide disulfide bond, too; see, for example, the EPO mimetic peptides in Table 1. A few examples of intrapeptide disulfide-linked peptides are specified in the Table. Any of these peptides can be derived as described herein, and a few derivative examples are provided in the Table. For derivatives in which the carboxyl terminus can be crowned with an amino group, the coronation amino group is shown as -NH2. For derivatives in which the amino acid residues are replaced by different portions to amino acid residues, substitutions are denoted by a d, which means any of the portions known in the art, for example, as described in Bhatnagar et al. (1996), J. Med. Chem. 39: 3814-9 and Cuthbertson et al. (1997), J. Med. Chem. 40: 2876-82, which are fully incorporated by reference. The substituent J and the substituents Z (Z5, Z6, ... Z40), are defined in the patents of E.U.A. Nos. 5,608,035, 5,786,331 and 5,880,096, which are hereby incorporated by reference in their entirety. For the EPO mimetic sequences (Table 1), the substituents X2 to Xn and the integer "n" are as defined in WO 96/40772, which is incorporated fully as a reference. The residues appearing in bold are amino acids D, but they may optionally be amino acids L. All peptides are linked via peptide bonds unless otherwise indicated. The abbreviations are listed at the end of this specification. In the column "SEQ ID NO", "NR", it means that sequence listing is not required for the given sequence.
TABLE 1 Peptide mimetic sequences of EPO Sequence / structure SEQ ID NO: YXCXXGPXTWXCXP 1 YXCXXGPXTWXCXP-YXCXXGPXTWXCXP 1 YXCXXGPXTWXCXP -? - YXCXXGPXTWXCXP 1 YXCXXGPXTWXCXP -? - (1 YXCXXGPXTWXCXP -? - (a-amine) 1 GGTYSCHFGPLTWVCKPQGG 2 GGDYHCRMGPLTWVCKPLGG 3 GGVYACRMGPITWVCSPLGG 4 VGNY CHFGPITWVCRPGGG 5 GGLYLCRFGPVTWDCGYKGG 6 GGTYSCHFGPLTWVCKPQGG 7 GGTYSCHFGPLTWVCKPQGG-GGTYSCHFGPLTWVCKPQGG 7 GGTYSCHFGPLTWVCKPQGG -? - GGTYSCHFGPLTWVCKPQGG 7 GGTYSCHFGPLTWVCKPQGGSSK 8 GGTYSCHFGPLTWVCKPQGGSSK-GGTYSCHFGPLTWVCKPQGGSSK 8 GGTYSCHFGPLTWVCKPQGGSSK -? - GGTYSCHFGPLTWVCKPQGGSSK 8 GGTYSCHFGPLTWVCKPQGGSS (e-amine) K / ßA / GGTYSCHFGPLTWVCKPQGGSS (a-amine) 8 GGTYSCHFGPLTWVCKPQGGSSK (-? - biotin) 8 CX4X5GPX6TWX7C 9 GGTYSCHGPLTWVCKPQGG 10 VGNYMAHMGPITWVCRPGG 11 GGPHHVYACRMGPLTWIC 12 GGTYSCHFGPLTWVCKPQ 13 GGLYACHMGPMTWVCQPLRG 14 TIAQYICYMGPETWECRPSPKA 15 YSCHFGPLTWVCK 16 YCHFGPLTWVC 17 YX2X3 X4X5G PX 6TWX7X8 19 X-? YX2X3X4X5GPX6X7X8X9X? OX ?? twenty X1YX2CX4X5GPX6TWX7CX9X10X ?? twenty-one GGLYLCRFGPVTWDCGYKGG 22 'GGTYSCHFGPLTWVCKPQGG 23 VGNYMCHFGPITWVCRPGGG 24 GGVYACRMGPITWVCSPLGG 25 TIAQYICYMGPETWECRPSPKA 26 YSCHFGPLTWVCK 27 YCHFGPLTWVC 28 SCHFGPLTWVCK 29 (AX2) nX3X4X5GPX6TWX7X8 30 The biological activities of EPO are well known in the art. See, for example, Anagnostou A et al. Erythropoietin has a mitogenic and positive chemotactic effect on endothelial cells. Proceedings of the National Academy of Science (USA) 87: 5978-82 (1990); Fandrey J and Jelkman WE Interleukin 1 and tumor necrosis factor-alpha inhibit erythropoietin production in vitro. Annals of the New York Academy of Science 628: 250-5 (1991); Geissler K et al., Recombinant human erythropoietin: A multipotential hemopoietic growth factor in vivo and in vitro. Contrib. Nephrol. 87: 1-10 (1990); Gregory CJ Erythropoietin sensitivity as a differentiation marker in the hemopoietic system. Studies of three erythropoietic colony responses ¡n culture. Journal of Cellular Physiology 89: 289-301 (1976); Jelkman W et al Monokines inhibiting erythropoietin production in human hepatoma cultures and in isolated perfused rat kidneys. Life Sci. 50: 301-8 (1992); Kimata H et al Human recombinant erythropoietin directly stimulates B cell immunoglobulin production and proliferation in serum-free medium. Clinical and Experimental Immunology 85: 151-6 (1991); Kimata H et al Erythropoietin enhances immunoglobulin production and proliferation by human plasma cells in a serum-free medium. Clin. Immunology Immunopathol. 59: 495-501 (1991); Kimata H et al. Effect of recombinant human erythropoietin on human IgE production in vitro Clinical and Experimental Immunology 83: 483-7 (1991); Koury MJ and Bondurant MC Erythropoietin retards DNA breakdown and prevent programmed cell death in erythroid progenitor cells. Science 248: 378-81 (1990); Lim VS et al Effect of recombinant human erythropoietin on renal function in humans. Kidney International 37: 131-6 (1990); Mitjavila MT et al Autocrine stimulation by erythropoietin and autonomous growth of human erythroid leukemic cells in vitro. Journal of Clinical Investigation 88: 789-97 (1991); Andre M et al Performance of an immunoradiometric assay of erythropoietin and results for specimens from anemic and polycythemic patients. Clinical Chemistry 38: 758-63 (1992); Hankins WD et al. Erythropoietin-dependent and erythropoietin-producing cell lines. Implications for research and for leukemia therapy. Annals of the New York Academy of Science 554: 21-8 (1989); Kendall RGT et al Storage and preparation of samples for erythropoietin radioimmunoassay. Clin. Lab. Haematology 13: 189-96 (1991); Krumvieh D et al Comparison of relevant biological assays for the determination of biological active erythropoietin. Dev. Biol. Stand. 69: 15-22 (1988); Ma DD et al Assessment of an EIA for measuring human serum erythropoietin as compared with RIA and an in-vitro bioassay. British Journal of Haematology 80: 431-6 (1992); Noe G et al A sensitive sandwich ELISA for measuring erythropoietin in human serum British Journal of Haematology 80: 285-92 (1992); Pauly JU et al. Highly specific and highly sensitive enzyme immunoassays for antibodies to human interleukin 3 (IL3) and human erythropoietin (EPO) in serum. Behring Institui Mitteilungen 90: 112-25 (1991); Sakata S and Enoki Y Improved microbioassay for plasma erythropoietin based on CFU-E colony formation. Ann. Hematology 64: 224-30 (1992); Sanengen T et al Immunoreactive erythropoietin and erythropoiesis stimulating factor (s) in plasma from hypertransfused neonatal and adult mice. Studies with a radioimmunoassay and a cell culture assay for erythropoietin. Acta Physio. Scand. 135: 11-6 (1989); Widness JA et al A sensitive and specific erythropoietin immunoprecipitation assay: application to pharmacokinetic studies. Journal of Lab. Clin. Med. 119: 285-94 (1992); for an additional information, see also the individual cell lines used in the individual bioassays. Each of the above references is incorporated herein by reference in its entirety. The EPO can be tested using cell lines such as HCD57, NFS-60, TF-1 and UT-7, that respond to the factor. The activity of EPO can also be assessed in a colony formation assay, by determining the number of CFU-E of bone marrow cells. A different method of defection and complete differentiation is the quantification by RT-PCR of cytokines. A central mimetibody of the hinge region, EPO mimetic, or a specified portion or variant thereof, which partially or substantially preferably provides at least one biological activity of at least one protein or fragment, can be bound to the protein or ligand of the fragment and thus provide at least one activity that is mediated in a manner other than by binding the proiein to at least one ligand or receptacle of the propheine or from other dependent or pro-mediated mechanisms. As used in the present, the term "aclivity of the mimic body of the hinge region, mimic of the EPO", refers to a central mime-body of the hinge region, mimicking the EPO that may modulate or cause minus a protein 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 trial.
The ability of a mimei-body of the hinge region, mimic of the EPO or a portion or variant specified to provide at least one protein-dependent activity, is preferably assessed by at least one biological assay of the appropriate protein, such as is described herein and / or as is known in the art. A central human mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the invention, may be similar to any class (IgG, IgA, IgM, etc.) or isotype and may comprise at least a portion of a light chain kappa or lambda. In one embodiment, the central human mimetibody of the hinge region, EPO mimetic or a specified portion or variant, comprises a fragment or variable of the heavy chain of IgG, a hinge region, CH2 and CH3, for example, minus one of the isotypes, lgG1, lgG2, lgG3 or lgG4. At least one central mimetic of the hinge region, mimic of the EPO or a specified portion or variant of the invention, binds to at least one specified ligand, specific to at least one proiein, subunit, fragment, portion or any combination thereof. the same. The at least one mimic peptide of the EPO of at least one central mimei-body of the hinge region, EPO mimetic, a specified portion or variant of the present invention, may optionally be linked to at least one specified ligand 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 the coníiguous amino acids of the sequences selected from the group consisting of a protein ligand, as an EPO receptor or a portion thereof. Such antibodies can be prepared by linking several portions of Formula (1) of the mimetic body of the hinge, EPO mimetic region using known techniques, preparing and expressing at least one (i.e., one or more) nucleic acid molecules encoding the mimeiibody of the hinge region, mimic of EPO, using known techniques of recombinant DNA technology or using any other suitable method as chemical syn- thesis. The mimelibodies that bind to the ligands or receptors of human EPO and that comprise at least a portion that defines a variable region of heavy or light chain, can be prepared using suitable methods, such as the phage display (Kaísube, Y., et al., Int J Mol. Med, 1 (5): 863-868 (1998)) or methods employing transgenic animals, as is known in the art and / or as described in the present. The central mimei-body of the hinge region, EPO mimetic, a specified portion or variant, can be expressed using the coding nucleic acid or portion thereof in a suitable host cell. Preferably, such mimetibodies or fragments that bind to the ligand thereof, can bind to the ligands or receptors of human EPO with high affinity (eg, KD less than or equal to approximately 10"7 M) The amino acid sequences that are substantially the same as the sequences described herein, include sequences that comprise conservative amino acid substitutions, as well as deletions and / or amino acid insertions. to the replacement of a first amino acid by a second amino acid that has chemical and / or physical properties (eg, charge, sphericity, polarity, hydrophobicity / hydrophilicity), which are similar to those of the first amino acid.Conservative substitutions include the replacement of a amino acid for another within the following groups: lysine (K), arginine (R) and hisidine (H), aspartate (D) and gluíamaío (E); asparagine (N), gluíamina (Q), serina (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), cisiein (C) and glycine (G); F, W and Y; C, S and T.
Amino Acid Codes The amino acids that constitute the mimetibodies or the specified portions or variants of the present invention are often abbreviated. The designations of the amino acids can be indicated by the designation of the amino acid by its one-letter code, its code of other words, name, or nucleoid codons, 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), as presented in the Table.
TABLE 2 A central mimetibody of the hinge region, mimetic of the EPO or a specified portion or variant of the present invention, may include one or more substitutions, deletions or additions of amino acids, either from naïve mutations or human manipulation, as specified herein. Such sequences or others that may be used in the present invention, include, but are not limited to, the following sequences presented in Table 3, as further described in Figures 1-42 of the provisional application of E.U.A. 60 / 507,349, filed on 09/30/2003, fully incorporated by reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed on June 17, 2004, incorporated complete herewith as reference, with the corresponding SEQ ID NOS: 31-72. These Referred Figures 1-42 (SEQ ID NOS: 31-72), or Figures 1-41 of PCT US04 / 19783, show examples of heavy / light chain variable / heavy region sequences, armbands / subdomains and their subscripts. , portions of which may be used in the Ig-derived proteins of the present invention, as taught herein.
TABLE 3 Of course, the number of amino acid substitutions that an expert would make depends on many factors, including those described above. Generally speaking, the number of amino acid substitutions, insertions or deletions for at least one of a central mimetibody of the hinge region, EPO mimic, will not be greater 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 in I presented.
The following description of the components of a central mimetibody of the hinge region of the EPO of the present invention is based on the use of formula I of the present invention, ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of an N term of an immunoglobulin variable region, P is at least one bioactive peptide, L is at least one binding polypeptide, H is at least a portion of at least one hinge region of the Immunoglobulin, CH2 is at least a portion of a CH2 constant region of immunoglobulin, CH3 is at least a portion of an immunoglobulin CH3 constant region, m, n, o, p, q, rys are independently an integer enire 0 , 1 or 2 and 10, which mimic different types of immunoglobulin molecules, for example, non-exclusively IgG1, IgG2, IgG3, IgG4, IgA, IgM, IgD, IgE and the like, or any subclass thereof, or any combination thereof. In the central mimetibodies of the hinge region of the present invention, the optional VN terminal portion may comprise 1-20 amino acids of at least one heavy chain variable framework region 1 (FR1), for example, as presented in the Figures 1-9 (SEQ ID NOS: 31-39), or at least one LC variable region, for example, as presented in Figures 10-31 (SEQ ID NOS: 40-61), of the US provisional application 60 / 507,349, filed on 09/30/2003, fully incorporated as reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed on June 17, 2004, hereby incorporated herein by reference., including substitutions, deletions or insertions as shown in these Figures, with those of the preferred Figures 5, 6 and 8. Also preferred are the variable sequences comprising the sequence Q-X-Q. Portion P may comprise at least any epidermal peptide as known in the art or as described herein, and as non-exclusive, those shown in Table 1, SEQ ID NOS: 1-30, or as known in the art, or any combination or consensus sequence thereof, or any fusion protein thereof. The optional linker sequence can be any peptide linker as is known in the art. The preferred sequence includes any combination of G and S, for example, XrX2-X3-X4-Xn, where X may be G or S, and n may be 5-30. Non-limiting examples include, GS, GGGS, GSGGGS, GSGGGSGG, and the like. In the present invention, the CH1 portion is not used and a variable number of amino acids of the N-terminus of the hinge region is deleted, for example, as referred to in Figures 1-42 of the provisional application of E.U.A. 60 / 507,349, presented on 09/30/2003, fully incorporated by reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed in June 17, 2004, incorporated herein by reference in its entirety, and Table 3. The variable number of amino acids used for the central portion of the hinge region of a mimetibody of the present invention includes, but is not limited to, deletion. of any of 1, 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, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 , 50, 51, 52, 53, 54, 55, 56, 57 or 1-3, 2-5, 2-7, 2-8, 3-9, 4-10, 5-9, 5-10, 5 -15, 10-20, 2-30, 20-40, 10-50, or any range or value therein, of the N-terminal amino acids of at least one hinge region, for example, as presented in the Figures 32-40 of the provisional US application 60 / 507,349, filed on 09/30/2003, fully incorporated by reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed on June 17, 2004, incorporated completely herein as reference, or Table 3 above, for example, non-exclusively, the deletion of any or all of amino acids 99-101 to 105-157 of amino acids 99-105, 99-108, 99- 111, 99-112, 99-113, 99-114, 99-115, 99-119, 99-125, 99-128, 99-134, 99-140, 99-143, 99-149, 99-155 and 99-158 of Figures 32-40 of the US provisional application 60 / 507,349, filed on 09/30/2003, fully incorporated by reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed on June 17, 2004, incorporated completely herein as reference, which correspond to SEQ ID NOS: 62-70, including the susfituciones, insertions or deletions described in Figures 32-40 of the provisional application of E.U.A. 60 / 507,349, filed on 09/30/2003, fully incorporated by reference herein, corresponding to Figures 1-41 of PCT Application No. US04 / 19783, filed on June 17, 2004, incorporated in full in the present as a reference. In the preferred embodiments, a hinge region of the present invention includes a deletion of the N term from the hinge region to provide a hinge region that includes an hasfa deletion, but not including a Cys or hasfa residue, but not including a Cys-Pro-Xaa-Cys sequence. In a further preferred embodiment, each center sequence of the hinge region used in a central body of the hinge region of the present invention includes amino acids 109-113 or 112-113 of Figure 36 (SEQ ID NO: 66). (IgG1); 105-110 or 109-110 of Figure 37 (SEQ ID NO: 67) (IgG2); 111-160, 114-160, 120-160, 126-160, 129-160, 135-160, 141-160, 144-160, 150-160, 156-160 and 159-160 of Figure 38 (SEQ ID. NO: 68) (IgG3); or 106-110 or 109-110 of Figure 39 (SEQ ID NO: 69) (IgG4). The sequences CH2, CH3 and optional CH4 can be any human sequence suitable or comparable to human, for example, as presented in Figures 1-42 of the provisional application of E.U.A. 60 / 507,349, filed on 09/30/2003, filed as a reference herein, corresponding to Figures 1-41 of the PCT Application No. US04 / 19783, filed on June 17, 2004, fully incorporated herein by reference, and Table 3, or as is known in the art, or any combination or consensus sequence thereof, or any fusion thereof. The amino acids in a mimic member of the hinge region, mimic of the EPO or a specified portion or variant of the present invention, which are essential for function, can be identified by methods known in the art, such as silio-directed mugegenesis. or the alanine scanning mufagenesis (eg, Ausubel, supra, Chapters 8, 15; Cunningham and Wells, Science 244: 1081-1085 (1989)). The last procedure introduces unique alanine mulations in each residue in the molecule. The resulting mutant molecules are then tested for biological activity, such as, non-exclusively, at least one activity related to the protein, as specified in the present or as is known in the art. Those sites that are critical to the central region of the hinge region, mimic of EPO, or a specified portion or variant that binds, may also be identified by structural analyzes, such as crystallization, nuclear magnetic resonance, or photoaffinity-labeled (Smlth, et al., J. Mol. Biol. 224: 899-904 (1992) and de Vos, et al., Science 255: 306-312 (1992)). The mimetibodies or specified portions or variants of the present invention may comprise, as the portion P of Formula (I), non-exclusively, at least one portion, sequence or combination selected from 3 to all of at least one of SEQ ID NOS: 1-30. The Non-limiting variants that can improve or maintain at least one of the activities listed above include, but are not limited to, any of the above polypeptides, which further comprise at least one mutation corresponding to at least one substitution, insertion or deletion that does not affect Significantly, the appropriate biological activities or functions of the central mimetibody of the hinge region mimic EPO. A central mimetibody of the hinge region, EPO mimetic or a specified portion or variant may optionally comprise, at least a functional portion of at least one polypeptide such as the P portion of Formula (I), at least one of 90-100 % of SEQ ID NOS: 1-30. A central mimetibody of the hinge, mimetic region of the EPO may optionally further comprise an amino acid sequence for the P portion of Formula (I), selected from one or more of SEQ ID NOS: 1-30. In one embodiment, the amino acid sequence P or a portion thereof, has approximately 90-100% identity (i.e., 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any interval or value therein) with the corresponding amino acid sequence of the corresponding portion of at least one of SEQ ID NOS: 1-30. Preferably, 90-100% identity of the amino acid sequence (i.e., 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any range or value therein) , it is determined by using an appropriate computer algorithm, as is known in the art.
The mimetibodies or specified portions or variants of the present invention may comprise any number of contiguous amino acid residues of a central mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention, wherein the number is selects from the group of integers consisting of 10-100% of the number of contiguous residues in a central mimetibody of the hinge region, EPO mimic. Optionally, this subsequence of conical 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 of longitude, or any value or value therein. In addition, the number of sub-sequences may be any integer selected from the group consisting of 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. As those with experience will appreciate, the invention includes at least one mimei-body of the hinge region, mimic of the biologically active EPO or a specified portion or variant of the present invention. The mimeiibodies or biologically active specified portions or variants have a specific activity of 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 naphiva (nonsyneffective), endogenous or related prolein and protein inserted or fused known or specified portions or variants. The methods for testing and quantifying which measure enzymatic activity and specificity by substraction are well known to those skilled in the art. In another aspect, the invention relates to human mimeinibodies and fragments that bind to the ligand as described herein, which are modified by the covalent binding of an organic portion. Such modification can produce a central mimic body of the hinge region, mimic of EPO or a fragment that binds to the ligand with improved pharmacokinetic properties (e.g., increased serum half-life in vivo). The organic portion can be a linear or branched hydrophilic polymeric group, a fatty acid group or a fatty acid ester group. In the particular modalities, the hydrophilic polymeric group can have a molecular weight of about 800 to about 120,000 Dalfons, and can be a polyalkane glycol (e.g., polyethylene glycol (PEG), polypropylene glycol (PPG)), a carbohydrate polymer, an amino acid polymer or a polyvinyl pyrrolidone, and the fatty acid group or the fatty acid ester may comprise from about eight to about forty carbon atoms. The modified mimetibodies and fragments that bind to the ligand of the invention may comprise one or more organic portions that are covalently linked, directly or indirectly to the central mimetibody of the hinge region, mimetic of the EPO or to a specified portion or variant. Each organic portion that binds to a central mimetibody of the hinge region, EPO mimetic or a fragment that binds to the ligand of the invention, can be independently a hydrophilic polymeric group, a fatty acid group or a fatty acid ester group. As used herein, the term "fatty acid" embraces monocarboxylic acids and dicarboxylic acids. A "hydrophilic polymer group," as used herein, refers to an organic polymer that is more soluble in water than in octane. For example, polylysine is more soluble in water than in octane. Thus, a central mimetibody of the hinge, mimetic region of EPO modified by the covalent attachment of polylysine is encompassed by the invention. Hydrophilic polymers suitable for modifying the antibodies of the invention may be linear or branched and include, for example, polyalkane glycols (eg, PEG, monomethoxy-polyethylene glycol (mPEG), PPG and the like), carbohydrates (eg, dextran, cellulose, ollgosaccharides, polysaccharides and the like), polymers of hydrophilic amino acids (for example, polylysine, polyarginine, polyaspartate and the like), polyalkane oxides (for example, polyethylene oxide, polypropylene oxide and the like) and polyvinyl pyrrolidone. Preferably, the hydrophilic polymer that modifies the central mimetibody of the hinge, mimetic region of the EPO of the invention has a molecular weight of about 800 to about 150,000 Daltons as a separate molecular enume. For example, PEG250o, PEG5000, PEG750o, PEG9000, PEGioooo, PEG? 2500, PEG? 500o and PEG20ooo can be used, where the subscript is the average molecular weight of the polymer in Dalíons.
The hydrophilic polymeric group may be substituted with one to about six fatty acid alkyl or fatty acid ester groups. Hydrophilic polymers which are substituted with a fatty acid or fatty acid ester group can be prepared using 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 (eg, activated with N, N-carbonyl diimidazole) in a fatty acid or an ester of The fatty acid can be coupled to a hydroxyl group in a polymer. Fatty acids and fatty acid esters suitable for modifying the mimetibodies of the invention may be saturated or may contain one or more units of unsaturation. Fatty acids which are suitable for modifying the mimetibodies of the invention include, for example, n-dodecanoate (C-2, lauraio), n-ioradecanoaio (C 4, myriadio), n-octadecanoate (Cie, esilario). ), n-eicosanoaio (C2o, araquidato), n-docosanoato (C22, behenato), n-triacontanoato (C30), n-tetracontanoaío (C4o), cis-? 9-ocíadecanoado (Cis, oleato), all cis-? 5,8,11, 14-eicosatetraenoate (C20, arachidonate), octandioic acid, tetradecandioic acid, ociacdecandioic acid, docosandioic acid and the like. Suitable fatty acid esters include monoesters of dicarboxylic acids comprising a linear or branched lower alkyl group. The lower alkyl group may comprise from one to about twelve, preferably one to about six, carbon atoms.
The modified human molecules and fragments that bind to the ligand can be prepared by using suitable methods, such as by means of a reaction with a nail or more modifying agents. A "modifying agent" as used herein, refers to a suitable organic group (e.g., hydrophilic polymer, a fatty acid, a fatty acid ester) that comprises an activating group. An "activating group" is a chemical moiety or a functional group that can, under suitable conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group. For example, reactive groups with amine include lamellar electrophilic groups such as tosylate, mesylate, halo (chlorine, bromine, fluorine, iodine), esters of N-hydroxysuccinimidyl (NHS) and the like. Activating groups which can react with the thiols include, for example, maleimide, iodoacetyl, acryloyl, pyridyl disulfides, 5-phiol-2-nitrobenzoic acid (TNB-thiol) and the like. An aldehyde functional group can be coupled to molecules containing amine or hydrazide, and an azide group can react with a trivalent phosphorus group to form phosphoramidate or phosphorimide bonds. Suitable methods for producing acyivant groups in 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 linked directly to the organic group (for example, hydrophilic polymer, fatty acid, fatty acid ester), or through a linking portion, for example, a C1-C12 group divalenfe, wherein one or more carbon atoms can be replaced by a hetero-atom such as oxygen, nitrogen or sulfur. Suitable linker moieties include, for example, tetra-ethylene glycol, - (CH2) 3-, -NH- (CH2) 6-NH-, - (CH2) 2-NH- and -CH2-0-CH2-CH2-0-CH2 -CH2-0-CH-NH-. Modifying agents comprising a linking moiety, can be produced, for example, by reacting a mono-Boc-alkyldiamine (eg, mono-Boc-ethylenediamine, mono-Boc-diaminohexane) with a fatty acid in the presence of 1-amino acid. -3- (3-dimethylaminopropyl) carbodimide (EDC), to form an amide bond between the free amine and the carboxylaph of the fatty acid. The Boc group can be removed from the production by the trifluoroacetic acid (TFA) irradiation to expose a primary amine which can be coupled to the carboxyl group as described, or it can be reacted with maleic anhydride and the product resulfanized to cyclize to produce an acyl derivative derivative. maleimido of fatty acid. (See, for example, Thompson, et al., WO 92/16221, the teachings of which are incorporated herein by reference). The modified mimeiibodies of the invention can be produced by reacting a human mimei-body downstream of the hinge region, mimic of EPO or a fragment that binds to the ligand with a modifying organism. For example, the organic moieties can be attached to the mimeiibody of the hinge, mimic region of the EPO in a nonspecific manner using a modifying agent that reacts with an amine, for example, a NHS ester of PEG. The mimeficuerpos modified humans or ligand-binding fragments can also be prepared by reducing disulfide bonds (eg, in-chain disulfide bonds) of a mimic body of the hinge region, mimic of EPO, or a fragment that binds to the ligand. The central mimetibody of the hinge region, mimetic of the reduced EPO or a fragment that binds to the ligand can then be reacted with a modifying agent that reacts with an iole, to produce the central mimei-body of the hinge region, mimetic of the EPO modified of the invention. Modified human mimefibodies and ligand-binding fragments comprising an organic portion that binds to specific sites of a miter-body member of the hinge region, EPO mimetic or a specified portion or variant of the present invention can be prepared using suitable methods, such as reverse proieolysis (Fisch et al., Bioconjugate Chem., 3: 147-153 (1992), Werlen et al., Bioconjugate Chem., 5: 411-417 (1994); Kumaran et al., Protein Sci. 6 (10): 2233-2241 (1997); Itoh et al., Bioorg. Chem., 2, 4 (1): 59-68 (1996); Capellas et al., Biotechnol. Bioeng., 56 (4): 456-463 (1997)), and the methods described in Hermanson, GT, Bioconjugate Techniques, Academic Press: San Diego, CA (1996).
Compositions of Centeral Mammary Moieties of the Hinge Region, EPO mimics The present invention also provides at least one composition of a central mimetic of the hinge region, mimetic of EPO or a specified portion or variant comprising at least one, at least two, at least three, at least four, at least five, at least six or more mimefibodies or specified portions or variants thereof, as described in present and / or as is known in the art, which are provided in a composition, mixture or non-natural form. Such percentages of the composition are by weight, volume, concentration, molarity or molality as solutions, mixtures, suspensions, emulsions or liquid or dry colloids, as is known in the art or as described herein. Such compositions may comprise 0.00001-99.9999 percent by weight, volume, molarity concentration or molality as liquid, gas or dry solutions, mixtures, suspension, emulsions or colloids, as is known in the art or as described herein, at any interval or value therein, as, non-exclusively 0.00001, 0.00003, 0.00005, 0.00009, 0.0001, 0.0003, 0.0005, 0.0009, 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05, 0.09 0.1, 0.2, 0.3, 0.4, 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, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.3, 4.5, 4.6, 4.7, 4.8, 4.9, 5, 6, 7, 8 , 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7 , 99.8, 99.9%. Such compositions of the present invention include, but are not limited to, 0.00001-100 mg / ml and / or 0.00001-100 mg / g.
The composition may optionally further comprise an effective amount of at least one compound or protein selected from at least one of an amphetamine drug, a cardiovascular system drug (CV), a drug for the central nervous system (CNS), a drug for the autonomic nervous system (ANS), a drug for the respiratory tract, a drug for the gastrointestinal tract (Gl), a hormonal drug, a drug for the balance of fluids or electrolyte, a hematological drug, an antineoplastic drug, a drug for immunomodulation, an ophthalmic, otic or nasal drug, a topical drug, a nutritive drug or the like. Such drugs are well known in the art, including formulations, indications, dosage and administration for each presented herein (see, eg, Nursing 2001 Handbook of Drugs, 21st edition, Springhouse Corp., Springhouse, PA, 2001; Health Professional's Drug Guide 2001, ed., Shannon, Wilson, Stang, Prenfice-Hall, Inc, Upper Saddie River, NJ, PharmcoIherapy Handbook, Wells ef al., Ed., Appleton &Lange, Slamford, CT, each! Completed in the presence as reference). The antineoplastic drug may be at least one selected from amoebicides or at least one of antiprozozoa, anthelmintic, amphi- mycic, antimalarial, antimicrobial or at least one anilio- prolic, aminoglycosides, penicillins, cephalosporins, tel- cyclines, sulfonamides, fluoroquinolones, antivirals, amphi- infectious macrolides, anti-infectives. miscellaneous The drug for the CV may be at least one selected from Noiropic, antiarrhyme, antianginal, antihypertensive, anti-lipemic, miscellaneous cardiovascular drugs. The drug for the CNS may be at least one selected from non-narcotic analgesics or at least one selected from antipyretics, nonsteroidal anti-inflammatory drugs, narcotic drugs or at least one opioid analgesic, sedative hypnotics, anticonvulsants, antidepressants, antianxiety drugs, animimicos, sphimulanis. central nervous system, antiparkinsonians, miscellaneous drugs for the central nervous system. The drug for ANS may be at least one selected from cholinergic (parasympathomimetic) blockers, anticholinergic, adrenergic (simpalomiméíicos), adrenergic (sympatholytic), relaxants of the skeleíico muscle, neuromuscular blockers. The drug for the respiratory tract may be at least one selected from antihisphamines, bronchodilators, expectorants or at least one antifeedant, miscellaneous respiratory drugs. The drug for the Ig Gl can be at least one selected from amino acids or at least one of absorbents or at least one of amphiphillenol, digestive enzymes or at least one solubilizer of gallstones, antidiarrheals, laxatives, antiemetics, antiulcer drugs. The hormone drug may be at least one selected from corticosteroids, androgens or at least one anabolic spheroid, esogenous or at least one progesina, gonadotropin, antidiabetic drugs or at least one glucagon, thyroid hormones, thyroid hormone antagonists, pituitary hormones , drugs similar to the paraíiroides. The drug for balance Fluids and electrolytes can be at least one selected from diuretics, electrolytes or at least one replacement solution, acidifying or at least one alkalinizing. The hematological drug can be at least one selected from hemaynics, anicocoagulans, blood derivatives, thrombolytic enzymes. The antineoplastic agents may be at least one selected from alkylating agents, antimicrobial agents, antineoplastic agents, antineoplastic agents that alter the hormonal balance, miscellaneous antineoplastic agents. The drug for immunomodulation can be at least one selected from immunosuppressants, vaccines or at least one dioxoid, antioxyxin, or at least one amino acid, immune sera, biological response modifiers. Ophthalmic drugs, otic and nasal can be at least one selected from ophthalmic antiinfectives, ophthalmic antiinflammatories, miotics, midriáíicos, ophthalmic vasoconslrictores miscellaneous otic, nasal drugs. The topical drug may be at least one selected from local anti-infectives, scabicides or at least one pediculicide, topical corticosteroids. The nutritive drug can be at least one selected from vitamins, minerals or calories. See, for example, the content of the Nursing 2001 Drug Handbook, supra. The at least one amoebicide or antiprozozoate may be at least one selected from aiovaquone, chloroquine hydrochloride, chloroquine phosphate, meironidazole, meironidazole hydrochloride, pentamidine derivative. The at least one anylhelminic can be at least one selected from mebendazole, pyrantel pamoate, isobendazole. The least an antimicrobial can be at least one selected from amphocyeric B, amphoprotein B cholesteryl sulfate complex, lipid anfolericin B complex, liposomal amphotericin B, fluconazole, flucyosin, micro-size griseofulvin, ultramicro-sized griseofulvin, iiraconazole, quenoconazole, nisiaine, hydrochloride íerbinafina. The at least one antimalarial can be at least one selected from chloroquine hydrochloride, chloroquine phosphate, doxycycline, hydroxychloroquine sulfate, mefloquine hydrochloride, primaquine phosphate, pyrimethamine, pyrimethylamine with sulfadoxine. The at least one antituberculous or anilileproic acid can be at least one selected from clofazimine, cycloserine, dapsone, ethylbacillin hydrochloride, isoniazid, prazrazinemide, rifabutin, rifampin, rifapentine, streptomycin sulfate. The at least one aminoglycoside can be at least one selected from amikacin sulfate, geniamycin sulfate, neomycin sulfate, streptomycin sulfate, and бbramycin sulfate. The at least one penicillin can be at least one selected from amoxcillin / pofasium clavulanna, amoxicillin hydrolyzate, ampicillin, ampicillin sodium, ampicillin hydrolyzate, sodium ampicillin / sodium sulbacid, cloxacillin sodium, dicloxacillin sodium, sodium mezlocillin, sodium nafcillin, oxacillin sodium, penicillin G benzathine, penicillin G potassium, penicillin G procaine, penicillin G sodium, penicillin V potassium, piperacillin sodium, piperacillin sodium / iozobaciram sodium, disodium ticarcillin, disodium ticarcillin / polasium davulanate. The at least one cephalosporin can be at least one selected from at least one of cefaclor, cefadroxil, cefazolin sodium, cefdinir, hydrochloride of cefepime, cefixime, sodium cefmelazole, cefonic acid sodium, cefoperazone sodium, cefoximax sodium, cefotein disodium, cefoxycin sodium, cefpodoxime proxeil, cefprozil, ceftazidime, ceftibuiene, ceftizoxime sodium, ceftriaxone sodium, cefuroxime axetil, cefuroxime sodium, cephalexin hydrochloride, cephalexin monohydrate, cephradine, loracarbef . The at least one tetracycline may be at least one selected from demeclocycline hydrochloride, calcium doxycycline, doxycycline hyclate, doxycycline hydrochloride, doxycycline monohydrate, minocidine hydrochloride, teracycline hydrochloride. The at least one sulfonamide can be at least one selected from cotrimoxazole, sulfadiazine, sulfamethoxazole, sulfisoxazole, sulfisoxazole acelyl. The at least one fluoroquinolone can be at least one selected from alacrofloxacin 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 mesylation of alacrofloxacin, 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, amanidead hydrochloride, amprenavir, cidofovir, delavirdine mesylate, didanosine, efavirenz, famciclovir, fomivirsen sodium, foscamef sodium, ganciclovir, indinavir sulfate, lamivudine , lamivudine / zidovudine, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, rimanadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valaciclovir hydrochloride, zalcitabine, zanamivir, zidovudine. The at least one macrolide anti-infective can be at least one selected from azithromycin, clarifromycin, dirithromycin, basic erythromycin, erythromycin esylaromycin, erifromycin eyilsuccinate, erythromycin labyrinthion, erythromycin stearate. The at least one miscellaneous additive may be at least one selected from aztreonam, bacifracin, sodium chloramphenicol succinal, clindamycin hydrochloride, clindamycin palm hydrochloride., dindamicine phosphate, imipenem and sodium cilastaine, meropenem, niírofuraníoína microcrisíales, niírofuraníoína microcrisíales, quinuprisíina / dalfoprisíina, spectinomycin hydrochloride, Irimefoprima, vancomycin hydrochloride. (See, for example, pp. 24-214 of Nursing 2001 Drug Handbook). The at least one inohropic can be at least one selected from the amnio- na, digoxin, milicone lacium. The at least one amino acid may be at least one selected from adenosine, amiodarone hydrochloride, atropine sulfate, bretyl tosilafo, diltiazem hydrochloride, disopyramide, disopyramide phosphate, esmolol hydrochloride, flecalnide acetamide, ibufillda fumarate, hydrochloride lldocaine, mexilefin hydrochloride, moricizine hydrochloride, phenoxine, phenoxynine sodium, procainamide hydrochloride, propafenone hydrochloride, propranolol hydrochloride, quinidine bisulfate, quinidine gluconate, quinidine polygalacyuronide, quinidine sulfate, soiolol, tocainide hydrochloride, hydrochloride of verapamil. The at least one antiangina can be at least one selected from besilado de Amlodipidine, amylum amyloid, bepridyl hydrochloride, diltiazem hydrochloride, isosorbide dinitrate, isosorbide mononitrate, nadolol, nicardipine hydrochloride, nifedipine, nitroglycerin, propranolol hydrochloride, verapamil, verapamil hydrochloride. The at least one antihypertensive agent may 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, diliiazem hydrochloride, doxazosin mesylate, enalaprilat, enalapril maleate, eprosartan mesylate, felodipine, fenoldopam mesylate, sodium fosinopril, guanabenz acetate, guanadrel sulfate, guanfacine hydrochloride, hydralazine hydrochloride, irbesartan, isradipine, labetalol hydrochloride, lisinopril, losarían of potassium, methyldopa, methyldopa hydrochloride, succinate of metoprolol, metoprolol tartrate, minoxidil, moexipril hydrochloride, nadolol, nicardipine hydrochloride, nifedipine, nisoldipine, sodium nitroprusside, penbuiolol sulfate, perindopril erbumine, phentolamine mesylate, pindolol, hydrochloride of prazo sina, propanoic hydrochloride, quinapril hydrochloride, ramipril, felmisartan, terazosin hydrochloride, imolol maleate, irarlapril, valsaran, verapamil hydrochloride. The at least one antilipemic can be at least one selected from calcium atorvasfatin, sodium cerivastain, cholesyramine, cholesipyl hydrochloride, fenofibrate (micronized), fluvastatin sodium, gemfibrozil, lovastatin, nlacin, pravasaline sodium, simvasiaine. The at least one miscellaneous drug for the CV may be at least one selected from abciximab, alprosadil, arbutamine hydrochloride, cilosiazole, clopidogrel bisulfate, dipyridamole, eptifibalide, midodrine hydrochloride, pentoxifylline, ticlopidine hydrochloride, and irofiban hydrochloride. (See, for example, pp. 215-336 of Nursing 2001 Drug Handbook). The at least one non-narcotic or antipyretic analgesic can be at least one selected from acetaminophen, aspirin, magnesium choline irrisalicylate, diflunisal, magnesium salicylate. The at least one non-spheroidal anti-inflammatory drug may be at least one selected from celecoxib, diclofenac, sodium diclofenac, eiolaco, calcium fenoprofen, flurbiprofen, ibuprofen, indomeiacin, indomethacin sodium hydrochloride, ketoprofen, ketorolac tromethamine, nabumeine, naproxen , naproxen sodium, oxaprozin, piroxicam, rofecoxib, sulindaco. The at least one narcotic or opioid analgesic can be at least one selected from alfentanil hydrochloride, buprenorphine hydrochloride, butorphanol hydroxide, codeine phosphate, codeine sulfate, fentanyl citrate, fentanyl transdermal system, transmucosal fentanyl, hydromorphone hydrochloride , meperidine hydrochloride, meladone hydrochloride, morphine hydrochloride, morphine sulfate, morphine urea, nalbuphine hydrochloride, oxycodone hydrochloride, oxycodone pecillin, oxymorphone hydrochloride, pentazocine hydrochloride, pentazocine hydrochloride and naloxone hydrochloride, lactate of pentazocine, propoxyphene hydrochloride, propoxyphene napsylate, remifentanil hydrochloride, sufenanilic acid, framadol hydrochloride. The at least hypnotic sedative can be at least one selected from doral hydrate, estazolam, flurazepam hydrochloride, pentobarbital, sodium pentobarbital, sodium phenobarbital, secobarbiil sodium, temazepam, iozozolam, zaleplon, zolpidem tartraio. The at least one aniliconvulsant can be at least one selected from acetazolamide sodium, carbamazepine, clonazepam, dorazepaie deipotassium, diazepam, divalproex sodium, ethosuximide, fosfenitoin sodium, gabapenlin, lamotrigine, magnesium sulfate, phenobarbifal, sodium phenobarbital, phenytoin, phenytoin sodium, Sodium phenophin (extended, primidone, iagabine hydrochloride, topiramate, sodium valproate, valprolco acid. The at least one antidepressant may be at least one selected from amyrylpyridine hydrochloride, amitripylline pamoate, amoxapine, bupropion hydrochloride, cyAalopram hydrobromide, clomipramine hydrochloride, desipramine hydrochloride, doxepin hydrochloride, fluoxelline hydrochloride, imipramine hydrochloride, mipramine pamoate, mirtazapine, nefazodone hydrochloride, nortriptyline hydrochloride, paroxetine hydrochloride, phenelzine sulfate, sertraline hydrochloride, sulpharyl of iarylcypromine, maleate of viripramine, venlafaxine hydrochloride. The at least one antianxiety drug can be at least one selected from alprazolam, buspirone hydrochloride, chlordiazepoxide, chlordiazepoxide hydrochloride, dipolamic clorazepate, diazepam, doxepin hydrochloride, hydroxycin hydroquinone, hydroxyzine hydrochloride, hydroxycin pamoate, lorazepam, mephrobamate, Midazolam hydrochloride, oxazepam. The at least one animocyclic drug can be at least one selected from chlorpromazine hydrochloride, clozapine, flufenacin decanoate, enaninate from fluefenacin, flufenacin hydrochloride, haloperidol, haloperidol decanoa, haloperidol alocap, loxapine hydrochloride, loxapine succinate, mesoridazine besilaine, molindone hydrochloride, olanzapine, perphenazine, pimozide, prochlorperazine, queiapia fumarate, risperidone, thioridazine hydrochloride, thiofixene, thiothixene hydrochloride, hydrochloride of urea fluoride. The at least one central nervous system stimulus can be at least one selected from amphetamine sulfate, caffeine, dextroamphetamine sulfate, doxapram hydrochloride, mefanfetamine hydrochloride, mephiphenidaflor hydrochloride, modafinil, pemoline, phentermine hydrochloride. The at least one antiparkinsonian may be at least one selected from amantadine hydrochloride, mesylation of benztropine, biperiden hydrochloride, biperiden's lactate, bromocriptine mesylate, carbidopa-levodopa, eniacapone, levodopa, pergolide mesylate, pramipexole dihydrochloride, ropinirole, selegiline hydrochloride, iocapona, hydrochloride of iohexyphenylidyl. The at least one miscellaneous drug of the central nervous system can be at least one selected from bupropion hydrochloride, donepezil hydrochloride, droperidol, fluvoxamine maleate, lithium carbonate, lithium chloride, narayriphenium hydrochloride, polacrilex nicoin, nicoinic transdermal system , propofol, benzoate of rizaíipiphen, hydrochloride of the monohydramide of sibuiramine, succinaio of sumafripía, hydrochloride of lacrine, zolmiíipipía. (See, for example, pp. 337-530 of Nursing 2001 Drug Handbook). The at least one cholinergic (for example, parasympathomimetic) may be at least one selected from bethanechol chloride, edrophonium, neosigmine bromide, neostigmine melilsulfalo, physostigmine salicylate, pyridostigmine bromide. The at least one anticolinergist can be at least one selected from sulfate of atropine, dicyclomine hydrochloride, glycopyrrolate, hyoscyamine, hyoscyamine sulfate, propantheline bromide, scopolamine, scopolamine builyl bromide, scopolamine hydrobromide. The at least one adrenergic (sympathomimetic) may be at least one selected from dobutamine hydrochloride, dopamine hydrochloride, meraminol bitartraph, norepinephrine biomarrow, phenylephrine hydrochloride, pseudoephedrine hydrochloride, pseudoephedrine sulfate. The at least one adrenergic blocker (sympatholytic) can be at least one selected from dihydroergotamine mesylate, ergotamine tartrate, meiiséridle maleate, propranolol hydrochloride. The at least one skeletal muscle relaxant may be at least one selected from baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine hydrochloride, sodium danirolene, meiocarbamol, Iizanidine hydrochloride. The at least one neuromuscular blocker can be at least one selected from airacurium besylate, besylate of cisairacurium, doxacurium chloride, mivacurium chloride, pancuronium bromide, pipecuronium bromide, rapacuronium bromide, rocuronium bromide, succinylcholine chloride, chloride of fubocurarine, vecuronium bromide. (See, for example, pp. 531-84 of Nursing 2001 Drug Handbook). The at least one anisophislamine can be at least one selected from brompheramine maleate, cetiricin hydrochloride, maleate of chlorpheniramine, clemastine fumarate, ciprohepidead hydrochloride, diphenhydramine hydrochloride, fexofenadine hydrochloride, loratadine, promethazine hydrochloride, promethacillus pheoclonide, triprolidine hydrochloride. The at least one bronchodilator may be at least one selected from albuterol, albufferol sulfate, aminophylline, afropin sulfate, ephedrine sulfate, epinephrine, epinephine bitartrate, epinephrine hydrochloride, ipratropium bromide, soproferenol, isoprorenol hydrochloride, sulfa of isoproierenol, levalbuireol hydrochloride, metaprorenol sulfate, oxyfilin, acetamide of pirbuferol, xinafoalo of salmeferol, sulfuric acid, iophylline. The at least one specimen or anililusive can be at least one selected from benzonatoate, codeine phosphate, codeine sulfate, dexframethraphan bromhydrate, diphenhydramine hydrochloride, guaifenesin, hydromorphone hydrochloride. The at least one respiratory miscellaneous drug can be at least one selected from acetylcysteine, beclomethasone dipropionate, beractan, budesonide, calfacium, cromolyn sodium, dornase alfa, sodium epoprosenol, flunisolide, fluticasone propionate, sodium moniluck, nedocromil sodium, palivizumab, aceionide of triamcinolone, zafirlukast, zileuíona. (See, for example, pp. 585-642 of Nursing 2001 Drug Handbook). The at least one anhydride, adsorbent or anilifluoride can be at least one selected from aluminum carbonate, aluminum hydroxide, calcium carbonate, magaldrate, magnesium hydroxide, magnesium oxide, simethicone, sodium bicarbonate. The at least one digestive enzyme or The solubility of gallstones can be at least one selected from pancreafina, pancrelipase, ursodiol. The at least one antidiarrhoeic can be at least one selected from atapulgyl, bismuth subsalicylate, polycarbophil sodium, diphenoxylachloride hydrochloride or atropine sulfate, loperamide, octreotide acetate, opium tincture, opium tincture (canforated). The at least one laxative can be at least one selected from bisocodyl, polycarbophil sodium, cascara sagrada (Rhamnus purshiana), aromatic liquid liquid of cascara sagrada, liquid extract of cascara sagrada, castor oil, docusaio cam, docusato sodium, glycerin, lacíulosa, magnesium citrate, magnesium hydroxide, magnesium sulfate, methylcellulose, mineral oil, polyethylene glycol or electrolyte solution, psyllium, senna, sodium phosphates. The at least one antieméfic may be at least one selected from chlorpromazine hydrochloride, dimenhydrinate, dolasetron mesylate, dronabinol, graniseiron hydrochloride, medicine chlorhydrate, metocloproamide hydrochloride, ondanseíron hydrochloride, perphenazine, prochlorperazine, prochlorperazine edisilate, maleate prochlorperazine, promethazine hydrochloride, scopolamine, tietylperazine maleate, trimeiobenzamide hydrochloride. The at least one aniliucera drug may be at least one selected from cimetidine, cimefidine hydrochloride, famotidine, lansoprazole, misoprostol, nizaidin, omeprazole, rabeprozol sodium, bismuth ranifidine citrate, raniidin hydrochloride, sucralpha. (See, for example, pp. 643-95 of the Nursing 2001 Drug Handbook). The at least one corticosteroid may be at least one selected from beiameiason, acetylase from beiamephase or sodium phosphamide of beíamefasona, sodium phosphatase of betamefasona, aceíaío of cortisone, dexamethasone, dexamethasone acetate, dexamethasone sodium, phosphate, fludrocortisone acetamide, hydrocortisone, hydrocortisone acetyl, hydrocortisone cipiona, sodium hydrocortisone phosphate, succinate of hydrocortisone sodium, methylprednisolone , methylprednisolone acetate, methylprednisolone sodium succinate, prednisolone, prednisolone acetate, prednisolone sodium phosphate, prednisolone tebutapho, prednisone, riamcinolone, friamcinolone aceylonide, friamcinolone diacephia. The at least one androgen or anabolic spheroid can be at least one selected from danazol, fluoximesyerone, meyiliesioserone, decanoafo from nandrolone, fenpropionalo from nandrolone, isoserone, testosterone cypionate, isosiaerone enanaria, isosterone propionate, testosterone transdermal system. The at least one estrogen or progestin can be at least one selected from esterified estrogens, estradiol, estradiol cypionate, estradiol / fransdermic system from noreindrone acellum, estradiol valerate, esogenous (conjugates), esipropylate, eicinyl sphradiol, eylinyl sphradiol and desogestrel Ethyl estradiol and diacephalion of efinodiol, ethynyl sphradiol and desogestrel, ethinyl estradiol and diacetate of ethinodiol, ethinyl estradiol and levonorgesfrel, ethinyl estradiol and noreindrone, ethynyl sphradiol and acetic acid noreindrone, ethynyl estradiol and norgestimaph, ethynyl estradiol and norgesphrel, ethynyl estradiol and norelindrone and acetyl and ferrous fumarate, levonorgesyral, acelaide of medroxyprogesterone, mesyranol and noreindrone, norelindrone, norethindrone acetyl, norgestrel, progesterone. The least a gonadropropin may be at least one selected from ganirelix acetyl, gonadorelin acetyl, hisyrelin acellum, menopropins. The at least one amino acid or glucagon 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, sodium liothyronine, lyotrix, thyroid. The at least one antagonist of the uroid hormone can be at least one selected from meimazole, pofasium iodide, poisonous iodide (saurated solution), propyl-trifouracil, radioactive iodine (sodium iodide I311), strong iodine solution. The at least one hormone of the pituitary gland may be at least one selected from corticotropin, cosyntropin, desmofresin acetyl, leuprolide acetate, repository corticotropin, somatrem, somatropin, vasopressin. The at least one drug similar to the parathyroid, may be at least one selected from calcifediol, calcitonin (human), calcitonin (salmon), calcitriol, dihydrotaquiserol, disodium eidronaio. (See, for example, pp. 696-796 of Nursing 2001 Drug Handbook). The at least one diuretic can be at least one selected from acezozolamide, sodium acezozolamide, amiloride hydrochloride, bumetanide, chlorthalidone, sodium ethacrylate, eiacrine acid, furosemide, hydrochlorothiazide, indapamide, manifol, mepholazone, spironolactone, ororsesemida,,, riamriamíeíeíereno,,,, urea. The at least one electrolyte or replacement solution can be at least one selected from calcium oil, calcium carbonate, calcium chloride, calcium calcium, calcium glutionale, calcium glucerate, calcium gluconate, calcium lactate, calcium phosphate (dibasic), calcium phosphate (tribasic), dextran (high molecular weight), dextran (low molecular weight), hetastarch, magnesium chloride, magnesium sulfate, potassium acetaium, potassium bicarbonate, potassium chloride, potassium gluconate, Ringer's injection, Ringer's injection (lactate), sodium chloride. The at least one acidifying or alkalizing agent can be at least one selected from sodium bicarbonate, sodium lactate, tromethamine. (See, for example, pp. 797-833 of Nursing 2001 Drug Handbook). The at least one hemaynic can be at least one selected from ferrous fumarate, ferrous gluconate, ferrous sulfate, ferrous sulfate (dry), iron dextran, iron sorbitol, polysaccharide-iron complex, sodium-gluconate complex. The at least one anticoagulant may be at least one selected from ardeparin sodium, dalteparin sodium, danaparoid sodium, enoxaparin sodium, calcium heparin, heparin sodium, warfarin sodium. The at least one blood derivative can be at least one selected from 5% albumin, 25% albumin, antihemophilic factor, anti-inhibitor-coagulanfe complex, anlifrombine III (human), facifor IX (human), factor IX complex, fractions of plasma protein. The at least one thrombolytic enzyme can be at least one selected from allylase, anistreplase, reteplase (recombinase), stryptokinase, urokinase. (See, for example, pp. 834-66 of Nursing 2001 Drug Handbook).
The at least one alkylating agent can be at least one selected from busulfan, carboplafin, carmusin, chlorambucil, cisplaine, cyclophosphamide, ifosfamide, lomusin, mechlorefamine hydrochloride, melphalan, melphalan hydrochloride, streptozocin, iomozolomide, iolepa. The at least one animethabolite may be at least one selected from capecitabine, cladribine, cytarabine, floxuridine, fludarabine phosphate, fluorouracil, hydroxyurea, mercaptopurine, methorexafo, methotrexate sodium, thioguanine. The at least one antineoplastic antibiotic can be at least one selected from bleomycin sulfate, dactinomycin, daunorubicin liposomal citrate, daunorubicin hydrochloride, doxorubicin hydrochloride, doxorubicin liposomal hydrochloride, epirubicin hydrochloride, idarubicin hydrochloride, miyomycin, penllosyatin, plicamycin , valrubicin. The at least one antineoplastic that alters the hormonal equilibrium can be at least one selected from anasyrozole, bicaluumamide, sodium pyramusine phosphate, exemesin, fluamide, goserelin acetyl, leirozole, leuprolide acetyl, megesyral acetyl, nilulamide, lamoxyphene, tesfolacyone, foremifen coli. The at least one miscellaneous anineoplastic may be at least one selected from asparaginase, Calmette-Guerin bacilli (BCG) (live intravesical), dacarbazine, doceiaxel, eyoposide, eosophoside phosphate, gemcyanabine hydrochloride, irinoic acid hydrochloride, mihoranium, hydrochloride mifoxanilone, paclitaxel, pegaspargase, porfimer sodium, procarbazine hydrochloride, riomarximab, nitroside, fopoiecane hydrochloride, irasyuzumab, Finoinoin, vinblastine sulfate, vincrisine sulfate, tartraio vinorelbine. (See, for example, pp. 867-963 of Nursing 2001 Drug Handbook). The at least one immunosuppressant can be at least one selected from azafioprine, basiliximab, cyclosporine, daclizumab, lymphocyte immune globulin, muromonab-CD3, mycophenolate mofopyloyl, mycophenolate hydrochloride mofopil, sirolimus, tacrolimus. The at least one vaccine or toxoid may be at least one selected from the BCG vaccine, cholera vaccine, diphtheria and telanic toxoldes (adsorbed), diphtheria and telanic loxoids and adsorbed acellular pertussis vaccine, diphtheria and telanic toxoids and vaccine for complete cell iosin ferina, Haemophilius b conjugate vaccines, A (inactivated) hepatitis A vaccine, B (recombinant) hepatic vaccines, trivalent influenza virus vaccine 1999-2000 of types A and B (purified surface antigen), vaccine for trivalent influenza virus 1999-2000 of types A and B (subvirion or purified subvirion), vaccine for trivalent influenza virus 1999-2000 types A and B (complete viron), vaccine for Japanese encephalitis virus (inactivated), vaccine for Lyme disease (recombinant OspA), measles and mumps and rubella virus vaccine (live), measles and measles virus vaccine and measles (live), vaccine for measles virus (live vaccine), polysaccharide vaccine for meningococcus, vaccine for mumps virus (live), vaccine for pest, vaccine for pneumococci (polyvalent), vaccine for the poliovirus (uncluttered), poliovirus vaccine (live, oral, ivalent), vaccine for rabies (adsorbed), rabies vaccine (human diploid cells), rubella and mumps virus vaccine (live), rubella virus vaccine (live, attenuated), leiinic toxoid (adsorbed), foxoid íeíánico (fluid), vaccine for typhoid (oral), vaccine for typhoid (parenteral), polysaccharide vaccine for typhoid Vi, vaccine for varicella virus, vaccine for yellow fever. The at least one antitoxin or antivenom may be at least one selected from the antivenom of the black widow spider, in antivenom for Cróíalos (polyvalent), diphtheria antitoxin (equine), anlivenene Micrurus fulvius). The at least one Immune serum can be at least one selected from the immune globulin immune to cytomegalovirus (inlyvenous), the immune globulin of hepatitis B (human), intramuscular immune globulin, immune globulin iniravenous, immune globulin of rabies (human), Invasive globulin immune to respiratory syncytial virus (human), immune globulin Rh0 (D) (human), intravenous globulin immune to Rh0 (D) (human), globulin immune to tetenes (human), globulin immune to varicella zoster. The at least one biological response modifier can be at least one selected from aldesleukin, epoetin alfa, filgrasim, glatiramer acetate for injection, interferon alfacon-1, inferferon alfa-2a (recombinanie), interferon alfa-2b (recombinanie) , interferon beta-1a, interferon beta-1 b (recombinant), interferon gamma-1b, hydrochloride of levamisole, oprelvekin, sargramosfim. (See, for example, pp. 964-1040 of Nursing 2001 Drug Handbook).
The at least one ophthalmic agent can be selected from bacitracin, chloramphenicol, ciprofloxacin hydrochloride, erythromycin, geniamycin sulfa, 0.3% ofloxacin, polymyxin B sulfate, 10% sodium sulfacefamide, 15% sodium sulfacetamide, 30% sodium sulfacetamide. , бbramycin, vidarabine. The at least one ophthalmic agent may be at least one selected from dexamethasone, dexamethasone sodium phosphate, 0.1% sodium diclofenac, fluorometholone, flurbiprofen sodium, ketorolac, arnomethamine, prednisolone acetamide (suspension), prednisolone sodium phosphate (solution). The at least one mlotic can be at least one selected from acetylcholine chloride, carbachol (intraocular), carbachol (topical), ecoiodofuran iodide, pilocarpine, pilocarpine hydrochloride, pilocarpine nitrate. The at least one mydriatic can be at least one selected from atropine sulfate, cyclopentolate hydrochloride, epinephrine hydrochloride, epinephryl borate, homatropine hydrobromide, phenylephrine hydrochloride, scopolamine hydrobromide, tropicamide. The at least one ophthalmic vasoconstrictor can be at least one selected from naphazoline hydrochloride, oximeiazoline hydrochloride, hydroxyurea hydrochloride. The at least one miscellaneous ophthalmic can be at least one selected from aphidlonidine hydrochloride, bexololol hydrochloride, brimonidine tartrate, carteolol hydrochloride, dipivefrin hydrochloride, dorzolamide hydrochloride, emedasin difumarate, sodium fluorescein, queioiifen fumarate, laianoprosi, levobunolol hydrochloride, melipranolol hydrochloride, sodium chloride (hypertonic), timolol maleate. The at least one Óíico can be at least a selected one of boric acid, carbamide peroxide, chloramphenicol, triethylamine polypeptide oleate condensate. The at least one nasal drug can be at least one selected from beclomethasone dipropionate, budesonide, ephedrine sulfate, epinephrine hydrochloride, flunisolide, fluficasone propionate, naphazoline hydrochloride, oximeiazoline hydrochloride, phenylephrine hydrochloride, fetrahydrozoline hydrochloride, acephonido of triamcinolone, xylometazoline hydrochloride. (See, for example, pp. 1041-97 of Nursing 2001 Drug Handbook). The at least one local anti-infective may be at least one selected from acyclovir, amphotericin B, azelaic acid cream, bacifracin, bumaconazole niitara, clindamycin phosphate, clofrimazole, econazole nitrate, erythromycin, gentamicin sulfate, quenoconazole, mafenide acetyl, metronidazole (topical), miconazole nitrate, mupirocin, hydrochloride naftifine, neomycin sulfa, nilrofurazone, nisiafine, piala sulfadiazine, terbinafine hydrochloride, terconazole, tetracycline hydrochloride, thioconazole, tolnaftate. The at least one scabicide or pediculicide may be at least one selected from crotamiton, lindane, permethrin, pyrethrins. The at least one topical corticosteroid may be at least one selected from beimameonasone dipropionase, belameiasone valerate, clobefasol propionate, desonido, desoximeiasone, dexamethasone, sodium dexamethasone phosphate, diflorasone diacetate, fluocinolone acetonide, fluocinonide, flurandrenolide, propionate, fluticasone, halcionide, hydrocortisone, hydrocortisone acetate, hydrocortisone buíirate, valerate of hydrocortisone, mometasone furoate, friamcinolone acetonide. (See, for example, pp. 1098-1136 of Nursing 2001 Drug Handbook). The at least one vitamin or mineral can be at least one selected from vitamin A, complex of vine B, cyanocobalamin, folic acid, hydroxocobalamin, calcium leucovorin, niacin, niacinamide, pyridoxine hydrochloride, riboflavin, iamin hydrochloride, vifamin C, vilamine D, cholecalciferol, ergocalciferol, analog of vitamin D, doxercalciferol, paricalciol, violin E, analogue of vine K, phytonadione, sodium fluoride, sodium fluoride (topical), elements in chromium, copper, iodine, manganese , selenium, zinc. The at least one caloric can be at least one selected from infusions of amino acids (chrysphalins), infusions of amino acids in dexrose, infusions of amino acids with electrolytes, infusions of amino acids with electrolytes in dexfrosa, infusions of amino acids for hepatic failure, infusions of amino acids for alia meiabolic tension, infusions of amino acids for renal failure, dextrose, fat emulsions, medium chain triglycerides. (See, for example, pp. 1137-63 of the Nursing 2001 Drug Handbook). The antibody or polypeptide compositions of the mimetic, central region of the hinge region of the EPO of the present invention may further comprise at least any suitable and / or effective amount of a composition or pharmaceutical composition comprising at least one proiein or antibody of the invention. mimeiibody of the hinge region, mimic of EPO to a cell, tissue, organ, animal or patient in the need for such modulation, tracing or therapy, optionally further comprising at least one selected from at least one TNF aniagonist (eg, non-exclusively, a chemical antagonist of the TNF protein, an antibody or monoclonal or polyclonal fragment of TNF) , a soluble TNF receptor (e.g., p55, p70 or p85) or a fragment, fusion polypeptides thereof, or a small molecule TNF antagonist, e.g., protein I or II that binds to TNF (TBP -1 or TBP-II nerelimonmab, infliximab, enterocept, CDP-571, CDP-870, afelimomab, lenercept and the like), an antirheumatic drug (for example, methotrexate, auranofin, aurothioglucose, azathioprine, etanercept, sodium gold, sulfate) of hydroxychloroquine, leflunomide, sulfasalcin), a muscle relaxant, a narcotic, a nonsteroidal inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial ( for example, aminoglycoside, an antimicrobial, an antiparasitic, an aniviviral, a carbapenem, a cephalosporin, a flurorquinolone, a macrolide, a penicillin, a sulfonamide, a tetracycline, animicrobial oíros), an amphisoria, a corticosteroid, an anabolic steroid, an agent related to diabetes, a mineral, a nutrient, a thyroid agent, a vilamine, a calcium-related hormone, an amphiarrheal, an anti-inflammatory, an anti-emetic, an anti-ulcer, a laxanie, an anticoagulant, an erythropoietin (eg, epoxyelin) alpha), a filgrasim (for example, G-CSF, Neupogen), a sargramostim (GM-CSF, Leucine), an immunization, an immunoglobulin, an immunosuppressant (for example, basiliximab, cyclosporine, daclizumab), a growth hormone, a hormone replacement drug, an estrogen receptor modulator, a mydriatic, a cycloplegic, an alkylating agent, an anti-metabolite, a mitotic inhibitor, a radiopharmaceutical, an antidepressant, an antimaniac agent, an antisychic, an anxiolytic , a hypnotic, a sympathomimetic, a stimulant, donepezil, tacrine, a medication for asthma, a befa agony, an inhaled spheroid, a leucoiriene inhibitor, a methylxanine, a cromolyn, an epinephrine or the like, dornase alfa (Pulmozyme), a cytokine or an anphonygosis of the cifocin. Non-limiting examples of such cytokines include, nonexclusively, any of IL-1 to IL-23. Suitable dosages are well known in the art. See, for example, Wells et al., Eds., Pharmacotherapy Handbook, 2nd Edition, Applefon and Lange, Stamford, CT (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, CA (2000), each of the references is incorporated completely in the present as reference. Such compositions may also include ioxin molecules that are linked, joined, co-formulated or coadministered with at least one antibody or polypeptide of the present invention. The toxin may optionally act to selectively destroy the cell or pathological tissue. The pathological cell can be a cancer cell or another cell. Such toxins can be, in a non-exclusive manner, a purified or recombinant toxin or a fragment of toxin comprising at least one functional cytotoxic domain of the toxin, for example, selected from at least one of castor, diphtheria toxin, poison toxin, or a bacterial toxin. The term toxin also includes endoioxins such as exotoxins produced by any mutant or natural recombinant bacteria or viruses, which can 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, enterotoxin labile with the heat of enterotoxigenic E. coli (LT), heat-stable enterotoxin (ST), Shigella cytotoxin, Aeromonas enterotoxins, toxin 1 of toxic shock syndrome ( TSST-1), staphylococcal enterotoxin A (SEA), B (SEB) or C (SEC), eneroeroxins of Esfrepiococos and the like. Such bacteria include, but are not limited to, strains of enteroioxygenic E. coli species (ETEC), sickle-hemorrhagic E. coli (e.g., serotype strains 0157: H7), species of Esiophylococcus (e.g., Staphylococcus aureus, Staphylococcus pyogenes). , Shigella species (eg, Shigella dysenteriae, Shigella flexneri, Sliigella boydii, and Shigella sonnei), Salmonella species (eg, Salmonella typhi, Salmonella cholera-suis, Salmonella enteritidis), Clostridium species (eg, Clostridium perfringens) , Clostridium difficile, Clostridium botulinum), Camphlobacter species (eg, Camphlobacter jejuni, Camphlobacter fetus), Heliobacter species (eg, Hellobacter pylori), Aeromonas species (eg, Aeromonas sobria, Aeromonas hydrophila, Aeromonas caviae), Pleisomonas shigelloides, Yersina enterocolitica, Vibrios species (for example, Vibrios cholerae, Vibrios parahenaolyticus), Klebsiella species, Pseudomonas a eruginous and Streptococci See, for example, Slein, ed., INTERNAL MEDICINE, 3rd ed., Pp. 1-13, Liííle, Brown and Co., Boston, (1990); Evans et al., Eds., Bacterial Infections of Humans: Epidemiology and Control, 2a. Ed., Pp 239-254, Plenum Medical Book Co., New York (1991); Mandell et al, Principies and Practice of Infectious Diseases, 3a. Ed., Churchill Livingstone, New York (1990); Berkow et al., Eds., The Merck Manual, 16th edition, Merck and Co., Rahway, N.J., 1992; Wood et al, FEMS Microbiology Immunology, 76: 121-134 (1991); Marrack et al., Science, 248: 705-711 (1990), the content of the references is fully incorporated herein by reference. The compositions of the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant of the present invention, may further comprise at least one of any suitable auxiliary, such as, but not limited to, a diluent, binder, siabilizanie, shock absorbers, salts, lipophilic solvents, preservatives, helpers or similar. Pharmaceutically acceptable auxiliaries are preferred. Non-limiting examples of, and methods for preparing such sterile solutions are well known in the art, such as, non-exclusively, Gennaro, Ed., Remington's Pharmaceutical Sciences, 18th 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 composition of the central mimetibody of the region of hinge, mimic of the EPO, as is well known in the art or as described herein. The excipients and pharmaceutical additives useful in the present composition include, but are not limited to, proteins, peptides, amino acids, lipids and carbohydrates (for example, sugars, including monosaccharides, di, tri, tetra and oligosaccharides; sugar derivatives, such as aldiioles, aldonic acids, spherified sugars and the like, and polysaccharides or sugar polymers), which may 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. The representative amino acid / mimetic components of the hinge region, mimic of EPO or a specified portion or variant, which also function in a buffer capacity, include alanine, glycine, arginine, betaine, histidine, glutamic acid, aspartic acid , cysteine, lysine, leucine, isoleucine, valine, methionine, phenylalanine, aspartame and the like. Another favorite 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; Y aldiols, such as mannitol, xylitol, maltiol, lacfilol, xylitol sorbitol (glucitol), myoinosilol and the like. Preferred carbohydrate excipients for use in the present invention are mannitol, trehalose and raffinose. The mimetic compositions of the hinge, mimic region of the EPO may also include a buffer or an agent that adjusts the pH; Typically, the buffer is a salt prepared from an acid or organic base. Representative buffers include organic acid salts such as citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid or phylic acid salts; Tris, hydrochloride of bromethamine or phosphate buffers. Preferred buffers to be used in the present compositions are organic acid salts as a salt. Additionally, compositions of the mimeiibody of the hinge region, EPO mimetic region or a specified portion or variant of the invention, may include polymeric excipients / additives such as polyvinylpyrrolidones, phycole (a polymeric sugar), dextray (e.g. cyclodextrins, such as 2-hydroxypropyl-β-cyclodexyrin), polyethylene glycols, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antialiasing agents, insensitivity agents (for example, polysorbabies such as "TWEEN 20" and "TWEEN 80"), lipids (eg example, phospholipids, fatty acids,), spheroids (e.g., cholesterol), and chelating agents (e.g., EDTA).
These and the additional known excipients and / or pharmaceutical additives suitable for use in the central mimetic compositions of the hinge region, EPO mimetic according to the invention, are known in the art, for example, as listed in " Remington: The Science &Practice of Pharmacy ", 19th ed., Williams & Williams, (1995), and in "Physician's Desk Reference," 52nd ed., Medical Economics, Montvale, NJ (1998), the descriptions of which are fully incorporated herein by reference. Preferred carrier or excipient materials are carbohydrates (e.g., saccharides and aldiioles) and buffers (e.g., chloro) or polymeric agents.
Formulations As indicated above, the invention provides stable formulations, which, preferably, include a suitable buffer with physiological saline or a salt chosen, as well as optional preserved solutions and formulations, containing a preservative, as well as conserved formulations for multiple suitable uses for pharmaceutical or veterinary use, comprising at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant in a pharmaceutically acceptable formulation. The 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, alcohol benzyl, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (eg, hexahydrate), alkyl paraben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent. Any suitable concentration can be used as known in the art, such as 0.001-5%, or any value or value therein, such as, not exclusively 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05 0.09, 0.1, 0.2, 0.3, 0.4, 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, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.3, 4.5, 4.6, 4.7, 4.8, 4.9 or any interval or value in the same. Non-limiting examples include, without conservator, 0.1-2% m-cresol (e.g., 0.2, 0.3, 0.4, 0.5, 0.9, 1.0%), 0.1-3% benzyl alcohol (eg, 0.5, 0.9, 1.1, 1.5, 1.9, 2.0, 2.5%), 0.001-0.5% thimerosal (eg, 0.005, 0.01), 0.001-2.0 % phenol (eg, 0.05, 0.25, 0.28, 0.5, 0.9, 1.0%), 0.0005-1.0% alkylparabens (eg, 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 similar. As indicated above, the invention provides an article of manufacture, which comprises packing a material and at least one vial comprising a solution of at least one less a core mimetibody of the hinge region, EPO mimetic or a portion or variant. specified, with prescribed shock absorbers and / or preservatives, optionally in an aqueous diluent, wherein the packaging material comprises a label indicating that the solution can be maintained for a period of 1, 2, 3, 4, 5, 6, 9, 12, 18, 20, and 24, 30 , 36, 40, 48, 54, 60, 66, 72 hours or greater. The invention further comprises an article of manufacture, comprising a packaging material, a first vial comprising at least one mimei-body of the hinge region, mimetic of lyophilized EPO or a specified portion or variant, and a second vial that it comprises an aqueous diluent of a prescribed buffer or preservative, wherein the packaging material comprises a label which indicates to the patient to reconstruct the at least one mimetic body of the hinge region, mimic of the EPO or a specified portion or variant in the diluent aqueous, to form a solution that can be maintained for a period of twenty-four hours or more. The at least one central mimetibody of the hinge region, mimic of the EPO or a specified portion or variant used in accordance with the present invention, may be produced by recombinant means, including preparations of mammalian or transgenic cells, or may be purified from other biological sources, as described herein or as known in the art. The identification of the amounts of at least one central mimei-body of the hinge region, EPO mimetic or a portion or variant specified in the product of the present invention, includes amounts that provide after reconstitution, whether it is a wet / dry system , concentrations of about 1.0 μg / ml to about 1000 mg / ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, for example, solution formulations will differ from the transdermal, pulmonary, and transmucosal patch methods or osmotic or micropump. Preferably, the aqueous diluent optionally further comprises a pharmaceutically acceptable preservative. Preferred preservatives include those selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkyl paraben (meilyl, ethyl, propyl, builyl and the like), benzalkonium chloride, Benzeoniium, sodium dehydroacetate and thimerosal, or mixtures thereof. The concentration of the preservative used in the formulation is a sufficient concentration to provide an antimicrobial effect. Such concentrations are dependent on the selected conservative and are easily determined by the expert. Other excipients, for example, agents for isofonicity, buffers, amphodoxies, preservative improvers, may optionally be added and preferably diluted. An agent for isohonicity, 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 pH range, such as from about pH 4 to about pH 10, and the preferred one ranges from about pH 5 to about pH 9, and a more preferred one ranges from about 6.0 to about 8.0. Preferably, the formulations of the present invention have a pH of about 6.8 and about 7.8. Preferred buffers include phosphate buffers, more preferably, sodium phosphate, particularly phosphate buffered saline (PBS). Other additives, such as pharmaceutically acceptable solubilizers such as Tween 20 (polyoxyethylene (20) sorbifan monolaurate), Tween 40 (polyoxyethylene (20) sorbifan monopalmitate), Tween 80 (polyoxyethylene (20) sorbitan monooleate), Pluronic F68 (copolymers of polyoxyethylene-polyoxypropylene block), and PEG (polyphenylene glycol) or non-ionic surfactants such as polysorbate 20 or 80 or poloxamer 184 or 188, Pluronic® polyols, other block copolymers, and ionic binders 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 a plastic container is used to administer the formulation. The presence of a pharmaceutically acceptable surfactant mimics the propensity to aggregate from the proiein. The formulations of the present invention can be prepared by a process comprising mixing at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant and a conservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkyl paraben (methyl, ethyl, propyl, butyl and the like), banzalkonium chloride, benzethonium chloride, sodium dehydroacety and thimerosal or mixtures thereof in an aqueous diluent. The mixing of at least one central mimetibody of the hinge region, mimetic of the EPO or a specified portion or variant and a preservative 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 central mimetibody of the hinge region, EPO mimetic or a specified portion or variant in a buffered solution, is combined with the desired preservative in a buffered solution in sufficient quantities to provide the proiein and the preservative at the desired concentrations. Variations of this procedure will be recognized by one of ordinary skill in the art. For example, the order in which the components are added, if additional additives are used, the temperature and the pH at which the formulation is prepared, are all factors that can be optimized for the concentration and the means of administration used. The claimed formulations can be provided to the patients as clear solutions or as double vials comprising a vial of at least one central mimetic of the hinge region, mimetic of lyophilized EPO or a specified portion or variant that is reconstituted with a second vial that contains water, a preservative and / or excipients, preferably a phosphate and / or serum buffer physiological and a salt chosen, in an aqueous diluent. A single vial with the solution or a dual vial that requires reconstitution can be re-hybridized multiple times and may be sufficient for a single cycle or for multiple cycles of patient irradiation, and may thus provide a more convenient treatment regimen than those currently available. . The claimed manufactured articles of manufacture are useful for administration for a period of immediately to twenty-four hours or more. Accordingly, the claimed manufacturing articles present, offer significant advantages to the patient. The formulations of the invention can optionally and safely be stored at ambient temperatures at about 2 to about 40 ° C and maintain their biological activity of the protein for extended periods of time, thus allowing a label on the package to indicate that the The solution can be maintained and / or used for a period of 6, 12, 18, 24, 36, 48, 72 or 96 hours or more. If the conserved diluent is used, the product may include use at least one of 1-12 months, half a year, one and a half and / or two years. The solutions of at least one central mimetibody of the hinge region, EPO mimetic or a portion or variant specified in the invention, can be prepared by a process comprising mixing at least one central mimetibody of the hinge region, mimetic of the EPO or a specified portion or variant in an aqueous diluent. The mixing is carried out using conventional methods of dissolution and mixing. To prepare a suitable diluent, for example, a measured amount of at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant in water or buffer, is combined in sufficient amounts to provide the protein and optionally a preservative or buffer at the desired concentrations. Variations of this procedure will be recognized by someone with ordinary skill in the art. For example, the order in which the components are added, if additional additives are used, the temperature and the pH at which the formulation is prepared, are all factors that can be optimized for the concentration and the means of administration used. The claimed products can be provided to the patients as transradrenal solutions or as double vials comprising a vial of at least one half metal body of the hinge region., mimetic of lyophilized EPO or a specified portion or variant that is reconstituted with a second vial containing the aqueous diluent. A single vial with the solution or a dual vial requiring reconstitution may be re-used multiple times and may be sufficient for a single cycle or for multiple cycles of the patient's schedule, and may thus provide a more convenient regimen than the currently available ones. . The claimed products can be provided indirectly to patients by providing pharmacies, clinics or other such institutions and facilities, transparent solutions or double vials which comprise at least one central mimetibody of the hinge region, mimetic of lyophilized EPO or a specified portion or variant that is reconstiuted with a second vial containing the aqueous diluent. The translucent solution in this case can be up to one liter or even larger in size, providing a large deposit from which smaller portions of the solution of the at least one central mimetibody of the hinge, mimetic region of the EPO or a portion or variant specified, one or multiple times, to transfer them to smaller vials and be provided by the pharmacy or clinic to their clients and / or patients. Recognized devices comprising these single-vial systems include those pen injector devices for the delivery of a solution, such as Humajecí®, NovoPen®, B-D®Pen, AuíoPen® and Opíimen®. Recognized devices comprising a dual system include those pen injector systems for reconstituting a lyophilized drug in a cartridge for delivery of the reconstituted solution, such as HumairoPen®. The products claimed present include a packaging material. The packaging material shall provide, in addition to the information required by the regulatory agencies, the conditions under which the product may be used. The packaging material of the present invention provides instructions to the patient to reconstruct the at least one mimei-body of the hinge region, mimic of the EPO or a specified portion or variant in the aqueous diluent to form a solution and use the solution for a period of 2-24 hours or longer for the product of two vials, wet / dry. For a single vial, the product in solution, the label indicates that such a solution can be used for a period of 2-24 hours or more. The claimed products present are useful for pharmaceutical use of the product in humans. The formulations of the present invention can be prepared by a process comprising mixing at least one mimetic body of the hinge region, mimic of the EPO or a specified portion or variant and a selected buffer, preferably a phosphate buffer containing serum physiological or a salt chosen. The mixing of at least one mimetic body of the hinge region, mimicking the EPO or a specified portion or variant and the buffer in 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 mimic body of the hinge region, mimetic of EPO or a specified portion or variant in water or buffer, is combined with the desired buffer in water, in sufficient amounts to provide the protein and buffer in the desired concentrations. Variations of this procedure will be recognized by one of ordinary skill in the art. For example, the order in which the components are added, if additional additives are used, the temperature and the pH at which the formulation, are all factors that can be optimized for the concentration and the means of diversification used. The claimed spiked or preserved formulations can be provided to patients as clear solutions or as double vials comprising a vial of at least one central mimetibody of the hinge region, lyophilized EPO mimic or a specified portion or variant that is reconstituted with a second vial containing a preservative or buffer and excipients in an aqueous diluent. A single vial with the solution or a dual vial that requires reconstitution can be re-used multiple times and may be sufficient for a single cycle or for multiple cycles of the patient's treatment, and may thus provide a more convenient regimen than the currently available ones. . At least one central mimetibody of the hinge region, mimetic of the EPO or a portion or variant specified in the stable or preserved formulations or solutions described herein, may be administered to a patient according to the present invention, via a variety of delivery methods, including SC or IM injection; transdermal, pulmonary, transmucosal, implant, osmotic pump, cartridge, micropump or other means appreciated by the expert, well known in the art.
Therapeutic Applications The present invention for antibodies also provides a method for modulating or irradiating anemia, in a cell, tissue, organ, animal or patient, including, but not limited to, at least one of any anemia, anemia related to the anemia. for cancer, anemia related to radiotherapy or chemotherapy, anemia related to the rash of a viral or bacterial infection, renal anemia, anemia of prematurity, anemia associated with pediatric and / or adult cancer, anemia associated with lymphoma, myeloma , multiple myeloma, anemia associated with AIDS, concomitant traisation for patients with or without donation of aufologo blood waiting for an elective surgery, preoperative and postoperative for surgery, donation or blood transfusion auíóloga, perioperaíorio handling, cyclic neutropenia or Kosímann syndrome (agranulocytosis congenital), end-stage renal disease, anemia associated Dialysis, chronic renal failure, primary hemopoietic diseases, such as congenital hypoplastic anemia, major falasemia, sickle cell disease, vasoocclusive complications of sickle cell disease. Furman et al., Pediatrics 1992; 90: 716-728, Goldberg Science. 1988; 242: 1412-1415; Paul et al., Exp Hematol. 1984; 12: 825-830; Erslev et al., Arch Infern Med. 1968; 122: 230-235; Ersley et al., Ann Clin Lab Sci. 1980; 10: 250-257; Jacobs ei al., Nafure. 1985; 313: 806-810; Lin ei al., Proc Nati Acad Sci USA. 1985; 82: 7580-7584; Law al., Proc Naíl Acad Sci USA. 1986; 83: 6920-6924; Goldwasser ef al., J Biol Chem. 1974; 249: 4202-4206; Eaves eí al., Blood. 1978; 52: 1196-1210; Sawyer went to., Blood. 1989; 74: 103-109; Winearls et al., Lancet. 1986; 2: 1175-1178; Eschbach et al., N Engl J Med. 1987; 316: 73-78; Eschbach et al., Ann Intern Med. 1989; 111: 992-1000, each reference is fully incorporated herein by reference. The antibodies of the present invention can also be used for non-renal forms of induced anemia, for example, by chronic infections, inflammatory processes, radiation therapy and cytosylactic drug therapy, and aleniative results have been reported in patients with non-renal anemia. . See, for example, Abis Rl and Rudnick SA Erythropoietin: evolving clinical applications. Experimental Hemaíology 19: 842-50 (1991); Graber SE and Kraníz SB Erythropoietin: biology and clinical use. Hemafology / Oncol. Clin. North Amer. 3: 369-400 (1989); Jelkman W and Gross AJ (eds) Eryíhropoietin. Springer, Berlin 1989; Koury MJ and Bondurant MC The molecular mechanism of erythropoietin action. European Journal of Biochemistry 210: 649-63 (1992); Krantz SB Eryíhropoietin. Blood 77: 419-34 (1991); Tabbara IA Eryíhropoietin. Biology and clinical applications. Archives of Infernal Medicine 153: 298-304 (1993), each fully incorporated herein by reference. The present invention also provides a method for modulating or irradiating an anemia or condition related to blood cells, in a cell, tissue, organ, animal or patient, wherein the anemia or condition related to the blood cell is associated with at least one, including, but not limited to, at least one related immune disease, cardiovascular disease, infectious, malignant and / or neurological disease. Such a method may optionally comprise administering an effective amount of at least one composition or pharmaceutical composition comprising at least one central mimei-body of the hinge region, EPO mimetic or a specified portion or variant to a cell, protein, organ, animal. or patient in need of modulation, therapy or therapy. The present invention also provides a method for modulating or bringing the cancer / infectious disease into a cell, organ, animal, or patient, including, but not limited to, at least one of an acute or chronic bacterial infection., acute and chronic parasitic or infectious processes, including bacterial, viral and mycotic infections, HIV infection / neuropathy due to HIV, meningitis, hepatitis, septic arthritis, peritonitis, pneumonia, epiglottis, e. coli 0157: h7, haemolytic uraemic syndrome / myxobolic purpura frombociíopénica, malaria, dengue haemorrhagic fever, leishmaniasis, leprosy, toxic shock syndrome, streptococcal myositis, gas gangrene, mycobacterial tuberculosis, intracellular avium mycobacteria, pneumocystis carinii pneumonia, disease pelvic inflammatory disease, orchitis / epididymiis, legionella, lyme disease, influenza A, epsi-barr virus, vital associated hemaphagocytic syndrome, aseptic encephalitis / meningifis and the like; (ii) leukemia, acute leukemia, acute lymphoblastic leukemia (ALL), ALL of B lymphocytes, T lymphocytes or FAB, acute myeloid leukemia (AML), chronic myelocytic leukemia (CML), chronic lymphocytic leukemia (CLL), hairy cell leukemia, myelodysplastic syndrome (MDS), lymphoma, Hodgkin's disease, malignant lymphoma, non-hodgkin's lymphoma , Burkitt's lymphoma, multiple myeloma, Kaposi's sarcoma, colorectal carcinoma, pancreatic carcinoma, nasopharyngeal carcinoma, malignant histiocytosis, paraneoplastic syndrome / hypercalcemia of malignancy, solid tumors, adenocarcinomas, sarcomas, malignant melanoma, and the like; or (iii) neurodegenerative diseases, multiple sclerosis, migraine headache, complex dementia due to AIDS, disease-causing diseases, such as multiple sclerosis and acute transverse myelitis; extrapyramidal and cerebellar disorders such as lesions of the corticospinal system; írasíomos of the basal ganglia or cerebelar írastornos; hyperkinetic motion frasíomos such as Huntingfon's Korea and senile korea; movement frasures induced by drugs, such as those induced by drugs that block CNS dopamine receptors; hypokinetic disorders of the movement, such as Parkinson's disease; Progressive Paralysis of the supranucleus; cerebellar esírucfurales injuries; spinocerebellar degenerations, such as spinal ataxia, Friedreich's ataxia, cerebellar cortical degenerations, degenerations of multiple systems (Mencel, Dejerine-Thomas, Shi-Drager and Machado-Joseph); sysiemic írasíornos (Refsum's disease, abeíalipoproíemia, ataxia, íelangiecíasia and mitochondrial írastomo of multiple systems); disorders of the demyelinating nucleus, such as multiple sclerosis, myelitis acute transverse; and motor unit disorders such as neurogenic muscular atrophies (degeneration of the anterior horn cell, such as amyloid lateral sclerosis, infanilic spinal muscular atrophy, and juvenile spinal muscular atrophy); Alzheimer disease; Down syndrome in middle age; diffuse disease of the Lewy body; Senile dementia of the Lewy body type; Wemicke-Korsakoff syndrome; chronic alcoholism; Creutzfeldí-Jakob disease; subacute sclerosing panencephalitis, Hallerrorden-Spaíz disease and pugilistic dementia and the like. Such a method may optionally comprise administering an effective amount of a composition or pharmaceutical composition comprising at least one antibody to the TNF or a specified portion or variant to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy. See, for example, the Merck Manual, 16th Edition, Merck & Company, Rahway, NJ (1992). Such a method may optionally comprise administering an effective amount of at least one composition or pharmaceutical composition comprising at least one mimetic body of the hinge region, mimic of EPO or a 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, less one of a cardiac shock syndrome, myocardial infarction, congestive heart failure, stroke, ischemic stroke, haemorrhage, arteriosclerosis, aerosol, diabetic aetiosclerotic disease, hypertension, arterial hypertension, renovascular hypertension, syncope, cardiovascular syphilis, heart failure, pulmonary heart, primary pulmonary hypertension, cardiac arrhythmias, ectopic atrial beats, atrial agitation, atrial fibrillation (sustained or paroxysmal), chaotic or multifocal atrial tachycardia, QRS tachycardia, regular arrhythmias, specific arrhythmias, ventricular fibrillation, His bundle arrhythmias, atrioventricular block, branched beam block, myocardial ischemic disorders, coronary artery disease, chest angina, myocardial infarction, cardiomyopathy, dilated congestive cardiomyopathy, resuscitative cardiomyopathy, vascular heart disease, endocarditis, pericardial disease, cardiac tumors, aortic and peripheral aneurysms, aortic dissection, inflammation of the aorta, occlusion of the abdominal aorta and its branches, peripheral vascular disorders, occlusive arterial disorders, peripheral aà © cersclerotic disease, Buerger's disease, functional disorders of the peripheral artery, Raynaud's phenomenon and disease, acrocyanosis, erymromelalgia, venous diseases, venous thrombosis, varicose veins, arteriovenous fistula, linfederma, lipedema , unstable angina, reperfusion injury, posibomba syndrome, reperfusion ischemia injury and the like. Such a method may optionally comprise administering an effective amount of a pharmaceutical composition or composition comprising at least one mimetic body of the hinge region, EPO mimic or a specified portion or variant to a cell, tissue, organ, animal or patient in need of modulation, therapy or therapy. Any method of the present invention may comprise administering an effective amount of a composition or pharmaceutical composition comprising at least one mimic body of the hinge region, EPO mimic or a specified portion or variant to a cell, protein, organ, animal. or patient in need of modulation, treatment or therapy. Such a method may further comprise, optionally, coadministration or combination therapy to bring about immune diseases, wherein the administration of at least one central mimetibody of the hinge region, EPO mimetic, a specified portion or variant thereof, comprises also administer, before, concurrently and / or afterwards, at least one selected from at least one TNF antagonist (eg, non-exclusively, an antibody or fragment of TNF, a soluble receptor or fragments of TNF, proteins of fusion, or a small-molecule TNF antagonist), an antirheumatic drug, a muscle relaxant, a narcotic, a nonsteroidal anti-inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial (for example, aminoglycoside, an antimicrobial agent, an antiparasitic agent, an antiviral agent, a carbapenem, cephalosporin, a flurorquinolone, a macrolide, a penicillin, a sulfonamide, an iron oxide, an antimicrobial agent), an anisoaraic, a corticosteroid, a anabolic steroid, a diabetes-related organism, a mineral, a nutrient, a thyroid agent, a vine, a calcium-related hormone, an antidiarrheal, an antiemetic, an anti-emetic, an antiulcer, a laxanie, an anicoagulanle, an erythropoietin (e.g., epoetin alfa), a filgrasfim (e.g., G-CSF, Neupogen), a sargramostim (GM-CSF, Leucine), an immunization, an immunoglobulin, an immunosuppressant (e.g., basiliximab, cyclosporin, daclizumab), a growth hormone, a drug for hormone replacement, a modulator of the esophageal receptor, a mydriatic, a cycloplegic, an alkylating agent, an animemiabolite, a mitotic inhibitor, a radiopharmaceutical, an antidepressant, an antimaniaco agent, an antisychic, a Anxiolytic, a hypnotic, a sympathomimetic, a stimulant, donepezil, an iacrine, a medication for asthma, a beta agonist, an inhaled spheroid, a leukophrenic inhibitor, a methylxanoline, a cromolyn, an epinephrine or analog, dornase alfa (Pulmozyme), a cifocin or a cycloocin antagonist. Suitable dosages are well known in the art. See, for example, Wells et al., Eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, CT (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, CA (2000), each of the references are fully incorporated herein by reference. The mimetibodies can also be used ex vivo, such as in an anilic marrow culture. Briefly, the bone marrow is removed from a patient before chemotherapy and treated with TPO and / or EPO, 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 accelerate marrow recovery. In addition, TPO, alone or in combination with the EPO and / or EPO mimetibodies, can be used for the ex vivo expansion of the marrow in the peripheral blood progenitor cells (PBPC) of the marrow. Before the chemotherapy treatment, the marrow can be stimulated with the germ cell factor (SCF) or G-CSF to release the initial progenitor cells in the bloodstream. These parents are optionally collected and concentrated from the peripheral blood and then treated in culture with TPO and 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 in high megakaryocyte cultures. density, which optionally, is then returned to the patient after high-dose chemotherapy. The doses of TPO for ex vivo bone marrow treatment will be in the inervator from 100 pg / ml to 10 ng / ml, preferably 500 pg / ml to 3 ng / ml. The doses of the mimetibodies will be equivalent in acidity to EPO 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 value or value therein. TNF antagonists suitable for the compositions, combination therapy, co-administration, devices and / or methods of the present invention (further comprising at least one antibody, specified portion or variant thereof, of the present invention), include, but are not limited to, anti-TNF antibodies, ligand-binding fragments thereof, and receptor molecules that specifically bind to TNF; compounds which prevent and / or inhibit the synthesis of TNF, the release of TNF or its action on target cells, such as ialidomide, iodide, phosphodiesierase inhibitors (eg, penioxyfilline and rolipram), adenosine receptor agonists A2b and adenosine A2b receptor enhancers; compounds that prevent and / or inhibit TNF receptor signaling, such as inhibitors of mitogen-activated protein kinase (MAP); compounds that block and / or inhibit the excision of TNF from the membrane, such as inhibitors of meyaloproininase; compounds that block and / or inhibit the activity of TNF, such as inhibitors of the enzyme that converts angiotensin (ACE) (eg, capíopril); and compounds that block and / or inhibit the production and / or synthesis of TNF, such as MAP kinase inhibitors. As used in the present, an "antibody to tumor necrosis factor", "TNF antibody", "TNFα antibody" or fragment and the like, decreases, blocks, inhibits, cancels or interferes with the activity of TNFα. viir, in silu and / or preferably in vivo. For example, a suitable human TNF antibody of the present invention can bind to TNFa and include amph-TNF antibodies, fragments that bind to the antigen thereof, and specified mutants or domains thereof that specifically bind to TNFa. An antibody or fragment of the appropriate TNF it can also decrease, block, annul, interfere, prevent and / or inhibit RNA, TNF-DNA, or protein synthesis, TNF release, TNF receptor signaling, TNF cleavage from the membrane, activity of TNF, the production and / or synthesis of TNF. The chimeric cA2 antibody consists of a variable region that binds to the antigen of the high-affinity, neutralizing mouse anti-human IgG1 antibody to the IgG1 antibody, designated as A2, and the constant regions of human IgG1, immunoglobulin kappa. The Fc region of human IgG1 improves the effector function of the allogeneic antibody, increases the half-life in circulating serum and decreases the immunogenicity of the antibody. The strength and specificity of the chimeric cA2 antibody epitope are derived from the variable region of the murine A2 antibody. In a particular embodiment, a preferred source for the nucleic acids encoding the variable region of murine antibody A2 is the hybridoma A2 cell line. Chimeric A2 (cA2) neutralizes the cyto-toxic effect of natural and recombinant human TNFα in a dose-dependent manner. Of the binding assays of the chimeric cA2 antibody and the recombinant human TNFα, the affinity constant of the chimeric cA2 antibody was calculated to be 1.04 × 10 10 M "1. Preferred methods for determining the specificity and affinity of the monoclonal antibody by inhibition Competitive, can be found in Harlow, et al., Antibodies: A Laboraory Manual, Cold Spring Harbor Laboraory Press, Cold Spring Harbor, New York, 1988; Colligan et al., eds., Currení Protocols in Immunology, Greene Publishing Assoc. and Wiley Inícscience, New York, (1992-2003); Kozbor went to., Immunol. Today, 4: 72-79 (1983); Ausubel et al., Eds. Currení Profocols in Molecular Biology, Wiley Interscience, New York (1987-2003); and Muller, Meth. Enzymol., 92: 589-601 (1983), references which are fully incorporated in the present reference. In a particular embodiment, the murine monoclonal antibody A2 is produced by a cell line designated c134A. The chimeric antibody cA2 is produced by a cell line designated c168A. Additional examples of anti-TNF monoclonal antibodies that can be used in the present invention are described in the art (see, for example, U.S. Patent No. 5,231, 024).; Moller, A. et al., Cytokine 2 (3): 162-169 (1990); Application of E.U.A. No. 07 / 943,852 (filed on September 11, 1992); Rathjen e al., International Publication No. WO 91/02078 (published February 21, 1991); Rubin et al., EPO Patent Publication No. 0 218 868 (published April 22, 1987); Yone et al., EPO Patent Publication No. 0 288 088 (October 26, 1988); Llang, et al., Biochem. Biophys. Res. Comm. 137: 847-854 (1986); Meager, et al., Hybridoma 6: 305-311 (1987); Fendly et al., Hybridoma 6: 359-369 (1987); Bringman, et al., Hybridoma 6: 489-507 (1987); and Hirai, et al., J. Immunol. Meth. 96: 57-62 (1987), references which are incorporated herein by reference in their entirety).
TNF Receptor Molecules The preferred TNF receptor molecules useful in the present invention are those that bind to alpha affinity to TNFα (see, for example, Feldmann et al., International Publication No. WO 92/07076 (published in April 30, 1992), Schall et al., Cell 61: 361-370 (1990), and Loetscher et al., Cell 61: 351-359 (1990), references which are incorporated herein by reference in full) and optionally have low immunogenicity. In particular, cell surface receptors of 55 kDa (p55 TNF-R) and 75 kDa (p75 TNF-R) TNF are useful in the present invention. Truncated forms of these receptors, comprising the extracellular domains (ECD) of the receptors or the functional portions thereof (see, for example, Corcoran et al., Eur. J. Biochem. 223: 831-840 (1994) ), are also useful in the present invention. Truncated forms of TNF receptors, comprising the ECD, have been detected in urine and serum as 30 kDa and 40 kDa TNFa inhibitory binding proteins (Engelmann, H. et al., J. Biol. Chem. 265: 1531-1536 (1990)). The multimeric molecules of the TNF receptor and the TNF immunoreceptor fusion molecules and the derivatives or fragments or portions thereof, are additional examples of the TNF receptor molecules that are useful in the methods and compositions of the present invention. . The TNF receptor molecules that can be used in the invention are characterized by their ability to treat patients for extended periods with a relief of symptoms of good to excellent and low toxicity. Low immunogenicity and / or affinity, as well as undefined properties, may contribute to the therapeutic results achieved. The 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 non-peptide linkers, such as polyethylene glycol (PEG). The multimeric molecules may further comprise a signal peptide of a secreted protein to direct the expression of the multimeric molecule. These multimeric molecules and the methods for their production have been described in the application of E.U.A. No. 08 / 437,533 (filed on May 9, 1995), the content of which is hereby incorporated herein by reference. The TNF immunoreceptor fusion molecules useful in the methods and compositions of the present invention comprise at least a portion of one or more immunoglobulin molecules and all or a functional portion of one or more TNF receptors. These immunoreceptor fusion molecules can be assembled as monomers or hetero or homomultimers. The immunoreceptor fusion molecules can also be monovalent or multivalent. An example of such TNF immunoreceptor fusion molecules is the 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. 21: 2883-2886 (19911; Ashkenazi et al., Proc. Nati, Acad. Sci. USA 88: 10535-10539 (1991); Peppel et al., J. Exp. Med. 174: 1483-1489 (1991); Kolls et al., Proc. Nati, Acad. Sci. USA 91: 215-219 (1994); Buíler et al., Cytokine 6 (6): 616-623 (1994); Baker et al., Eur. J Immunol., 24: 2040-2048 (1994), Beutler et al., U.S. Patent No. 5,447,851, and U.S. Application No. 08 / 442,133 (filed on May 16, 1995), each of the references is incorporated completely in the present as a reference). Methods for producing immunoreceptor fusion molecules can also be found in Capon et al., U.S. Pat. No. 5,116,964; Capón eí al., Patent of E.U.A. No. 5,225,538; and Capon et al., Nature 337: 525-531 (1989), references which are incorporated herein by reference in their entirety. An equivalentderivative, fragment or functional region of the TNF receptor molecule refers to the portion of the TNF receptor molecule, or the portion of the sequence of the TNF receptor molecule that encodes the TNF receptor molecule, which it is of sufficient size and the sequences to functionally resemble the TNF receptor molecules that can be used in the present invention (for example, it binds to TNFa with alias affinity and possesses low immunogenicity). A functional equivalent of the TNF receptor molecule also includes modified TNF receptor molecules, which functionally resemble the TNF receptor molecules that can be used in the present invention (e.g., binds to TNFa with high affinity and has low immunogenicity). For example, a functional equivalent of the TNF receptor molecule may contain a "SILENT" codon or one or more substitutions, deletions or additions of amino acids (for example, the substitution of an acidic amino acid for another acidic amino acid, or the substitution of a codon encoding the same hydrophobic amino acid or a different one by 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, for example, CopewifhCyfokines.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 may comprise a method for bringing a protein-mediated enzyme, which comprises administering an effective amount of a pharmaceutical composition or composition comprising at least one minor moiety of the hinge region, mimic of the EPO or a portion or variance specified to a cell, tissue, organ, animal or patient in need of modulation, irradiation or therapy. Such a method may optionally further comprise coadministration or combination therapy to irradiate immune diseases, wherein the administration of at least one half-body of the hinge region, mimic of the EPO, a specified portion or variant thereof, further comprises administering, simultaneously and / or concurrently, at least one selected from at least one of the other cytokines such as IL-3, -6 and -11; facíor germ cells; G-CSF and GM-CSF. Typically, the treatment of pathological conditions is effected by administering an effective amount or dosage of at least one central mimetic composition of the hinge region, EPO mimetic which totals, on average, a range of at least about 0.01 to 500 milligrams. of at least one mimeiibody of the hinge region, mimic of the EPO or a specified portion or variant / kilogram of patient per dose, and preferably at least about 0.1 to 100 milligrams of the central mimeiibody of the hinge region, mimic of the EPO or a specified portion or variant / kilogram of patient per single or multiple administration, depending on the specific activity contained in the composition. All in all, the effective serum concentration may comprise 0.1-5000 μg / ml of serum concentration by single or multiple administration. Suitable dosages are known to the medical practitioners and will, of course, depend on the particular disease state, the specific disease of the composition being administered and the particular patient being subjected to the treatment. In some cases, to achieve the desired therapeutic amount, it may be necessary to provide repeated administration, ie, repeated individual administrations of a particular dose verified or measured, wherein Individual administrations are repeated until the desired daily dose or effect is achieved. Preferred doses may optionally include 0.01, 0.02, 0.03, 0.04, 0.05. 0.06, 0.07, 0.08, 009, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 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 and / or 30 mg / kg / administration or any range, value or fraction of same, or to reach a serum concentration of 0.1, 0.5, 0.9, 1.0, 1.1, 1.2, 1.5, 1.9, 2.0, 2.5, 2.9, 3.0, 3.5, 3.9, 4.0, 4.5, 4.9, 5.0, 5.5, 5.9, 6.0, 6.5, 6.9, 7.0, 7.5, 7.9, 8.0, 8.5, 8.9, 9.0, 9.5, 9.9, 10, 10.5, 10.9, 11, 11.5, 11.9, 20, 12.5, 12.9, 13.0, 13.5, 13.9, 14.0, 14.5, 4.9, 5.0, 5.5, 5.9, 6.0, 6.5, 6.9, 7.0, 7.5, 7.9, 8.0, 8.5, 8.9, 9.0, 9.5, 9.9, 10, 10.5, 10.9, 11, 11.5, 11.9, 12, 12.5, 12.9, 13.0 , 13.5, 13.9, 14, 14.5, 15, 15.5, 15.9, 16, 16.5, 16.9, 17, 17.5, 17.9, 18, 18.5, 18.9, 19, 19.5, 19.9, 20, 20.5, 20.9, 21, 22, 23 , 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 96, 100, 200, 300, 400, 500 , 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500 and / or 5000 / ml of serum concentration by single or multiple administration, or any range, value or fraction thereof. Alternatively, the dosage administered may vary depending on 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 the event and the desired effect. Usually, a dosage of the active ingredient can be approximately 0.1 to 100 milligrams per kilogram of body weight. Ordinarily, 0.1 to 50, and preferably 0.1 to 10 milligrams per kilogram per administration or in a sustained release form, are effective to achieve the desired results. As a non-limiting example, the tracing of humans or animals may be provided as a one-time or periodic dosage of at least one core mimetic of the hinge region, EPO mimetic or a specified portion or variant of the present invention, 0.01 to 100 mg / kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 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, 45, 50, 60, 70, 80, 90 or 100 mg / kg, per day, in at least one of the day 1, 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, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40, or alternatively, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, or any combination thereof, using single doses, infusion or repeated. Dosage forms (composition) suitable for internal administration generally contain from about 0.0001 milligrams to about 500 milligrams of the active ingredient per unit or container. In these pharmaceutical compositions, the active ingredient will ordinarily be present in a quantity of about 0.5-95% by weight based on the total weight of the composition.
For parenteral administration, the central membrane of the hinge region, mimic of the EPO or a specified portion or variant, can be formulated as a solution, suspension, emulsion or lyophilized powder in association or separately provided, with a parenteral parenteral vehicle. acceptable. Examples of such vehicles are water, physiological saline, Ringer's solution, dextrose solution and 5% human serum albumin. Liposomes and non-aqueous vehicles, such as fixed oils, can also be used. The vehicle or freeze-dried 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 standard in this field.
Therapeutic Administration Many known and developed modes can be used in accordance with the present invention to administer effec- tively therapeutically effective amounts of at least one central mimefibody of the hinge region, EPO mimetic or a specified portion or variant according to the present invention. While pulmonary administration is used in the following description, other modes of administration may be used in accordance with the present invention with suitable results. A central mimetibody of the hinge, mimetic region of the EPO of the present invention may be supplied in a carrier, as a solution, emulsion, colloid or suspension or as a powder, using any of a variety of devices and methods owed for administration. by inhalation or other modes described in the present invention or known in the art.
Parenteral Formulations and Administration Formulations for parenteral administration may contain as common carriers water or sterile physiological 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 using an appropriate emulsifier or humidifier and a suspending agent, according to known methods. The agents for injection may be a non-toxic, non-orally administrable diluting agent, such as an aqueous solution or a sterile injectable solution or suspension in a solvent. As the vehicle or useful solvent, water, Ringer's solution, isotonic saline, etc., are allowed, such as ordinary solvency or suspension solvent, sterile non-volatile oil can be used. For these purposes, any kind of oil and non-volatile fatty acid can be used, including fatty oils or fatty acids naíurales or siníéíicos or semisiníéíicos; mono or di or naíural or synthetic or semisynthetic oligoglycerides. Parenteral administration is known in the art and includes, but is not limited to, conventional means of injections, a needle-free injection device pressurized with gas, as described in U.S. Pat. No. 5,851, 198, and a laser perforating device as described in the U.S. Patent. No. 5,839,446, fully incorporated herein by reference.
Alternate Delivery The invention is further related to the administration of at least one mimetic body of the hinge region, mimic of EPO or a portion or variant specified by parenteral, subcutaneous, intramuscular, intravenous, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal. The compositions of the protein, mimic body of the hinge region, EPO mimetic or a specified portion or variant, can be prepared for parenteral administration (subcutaneous, intramuscular or intravenous), particularly in the form of liquid solutions or suspensions.; for use in vaginal or rectal administration, particularly semi-solid forms such as creams and suppositories; for buccal or sublingual administration, particularly in the form of tablets or capsules; or intranasally, particularly in the form of powders, nasal drops or aerosols or certain agents; or transdermally, in particular in the form of a gel, ointment, lotion, suspension or delivery system. patch with chemical enhancers such as dimethyl sulfoxide to modify the structure of the skin or to increase the concentration of the drug in the transdermal patch (Junginger, et al., in "Drug Permeation Enhancement"; Hsieh, DS, Eds., p. 59-90 (Marcel Dekker, Inc. New York 1994, fully incorporated herein by reference), or with oxidizing agents that allow the application of formulations containing proteins and peptides in the skin (WO 98/53847), or application of electric fields to create transient irradiation such as electroporation, or to increase the mobility of drugs charged through the skin, such as iontophoresis, or ultrasound application such as sonophoresis (U.S. Patent Nos. 4,309,989 and 4,767,402). prior publications and patents are hereby incorporated by reference in their entirety).
Lung / Nasal Administration For pulmonary administration, preferably, at least one central mimetic composition of the hinge region, EPO mimetic or a specified portion or variant, is supplied in an effective particle size to reach the airways inferiors of the lung or sinuses. According to the invention, at least one central mimetic of the hinge region, EPO mimetic or a specified portion or variant can be delivered by any of a variety of inhalation or nasal devices known in the art for the administration of a therapeutic agent by inhalation. These devices are capable of depositing aerosolized formulations in the cavity of the sinuses or the alveoli of a patient, including metered dose inhalers, nebulizers, dry powder generators, sprinklers and the like. Other suitable devices for directing the pulmonary or nasal administration of the mimetic body of the hinge region, mimic of the EPO or a specified portion or variant, are also known in the art. Such devices may use formulations suitable for administration to distribute the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant in an aerosol. Such aerosols may comprise solutions (aqueous or non-aqueous tannins) or solid particles. Metered-dose inhalers, such as the Ventolin® metered-dose inhaler, typically use a propellant gas and require activation during inspiration (see, for example, WO 94/16970, WO 98/35888). Dry powder inhalers such as Turbuhaler ™ (Asirá), Rofahaler® (Glaxo), Diskus® (Glaxo), Spiros ™ inhaler (Dura), devices marketed by Inhale Therapeuíics, and the Spinhaler® powder inhaler (Fisons), use the respiration of a mixed powder (US 4668218 Assira, EP 237507 Assira, WO 97/25086 Glaxo, WO 94/08552 Dura, US 5458135 Inhale, WO 94/06498 Fisons, incorporated herein by reference in its entirety). Nebulizers such as AERx ™ Aradigm, the Ultravent® nebulizer (Mallinckrodt), and the Acom II® nebulizer (Marquest Medical Products) (US 5404871 Aradigm, WO 97/22376), references above they are fully incorporated herein by reference, produce aerosols of 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 representative of specific devices suitable for the practice of this invention, and are not intended to limit the scope of the invention. Preferably, a composition comprising at least one central mimic body of the hinge region, EPO mimetic or a specified portion or variant is delivered by a dry powder inhaler or a spray. There are several desirable characteristics of an inhalation device for the administration of at least one central membranum of the hinge region, EPO mimetic or a specified portion or variant of the present invention. For example, the delivery by the inhalation device is, in an advantageous, reliable, reproducible and accurate manner. The inhalation device can optionally supply small dry particles, for example, less than about 10 μm, preferably about 1-5 μm, for good breathability.
Administration of compositions of the central mimei-body of the hinge region, EPO mimetic or a portion or variant specified as a spray A spray comprising a composition of a central mimetibody of the hinge region, mimic of the EPO or a portion or specified variant, may be produced by passing a suspension or solution of at least one mimei-body of the hinge region, EPO mimetic or a specified portion or variant through a nozzle under pressure. The size and configuration of the nozzle, the applied pressure and the speed of the liquid feed can be chosen to achieve the desired output and particle size. An electro-vacuum may be produced, for example, by an electric field in connection with a capillary or nozzle feed. Advantageously, the particles of at least one protein of a composition of the central mimetibody of the hinge region, mimic of the EPO or a specified portion or variant, delivered by a sprayer, have a particle size of less than about 10 μm, preferably in the range of about 1 μm to about 5 μm, and more preferably about 2 μm to about 3 μm. Formulations of at least one protein of a composition of a central mimetibody of the hinge region, mimic of EPO or a specified portion or variant suitable for use with a sprayer, typically includes a protein of a composition of a mimetibody. central of the hinge region, mimetic of the EPO or a specified portion or variant in an aqueous solution at a concentration of about 1 mg to about 20 mg of at least one prolein of a composition of a mimei-body of the hinge region, EPO mimic or a portion or variance specified per ml of solution. The formulation may include agents such as an excipient, a buffer, an agent for isotonicity, a preservative, a surfactant and, preferably, zinc. The formulation may also include an excipient or agent for the stabilization of the protein of the mimei-body composition of the hinge region, EPO mimetic or a specified portion or variant, such as a buffer, a reducing agent, a protein bulk or a carbohydrate. Bulk proteins useful in the formulation of the composition proteins of the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant include albumin, protamine or the like. Typical carbohydrates useful in the formulation of proteins of the composition of the central mimetibody of the hinge region, mimetic of EPO or a specified portion or variant include sucrose, mannitol, lactose, trehalose, glucose or the like. The protein formulation of the mimeiibody composition of the hinge region, mimic of the EPO or a specified portion or variant may also include a pentioscintive, which may reduce or prevent protein-induced surface aggregation of the protein. composition of the mimeiibody of the hinge region, mimic of the EPO or a portion or specified variant, caused by the atomization of the solution when forming an aerosol. Various conventional surfactants can be employed, such as esters of chicken polyethylene fatty acids and alcohols and esters of polyoxyethylene sorbitol fatty acid. The amounts will generally vary between 0.001 and 14% by weight of the formulation. Especially preferred surfactants for the purposes of this invention are the polyoxyethylene sorbitan monooleate, polysorbate 80, polysorbate 20, or the like. Additional agents known in the art for the formulation of a prolein such as the mimeiibodies, or specified portions or variants, may also be included in the formulation.
Administration of the compositions of the central mimetibody of the hinge region, mimetic of the EPO or a portion or variant specified by means of a nebulizer The proiein of the composition of the mimeiibody of the hinge region, mimetic of the EPO or a specified portion or variant it can be administered by a nebulizer, such as a jet nebulizer or an ulysonic nebulizer. Typically, in a jet nebulizer, a source of compressed air is used to create a jet of air at a velocity through a hole. As the gas expands beyond the nozzle, a low pressure region is created, which exfers a solution of a protein from the composition of the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant. through a capillary tube connected to a liquid reservoir. The liquid stream of the capillary tube is cut into unstable filaments and gofas as it leaves the tube, creating the aerosol. A range of configurations, flow rates and types of baffles can be used to achieve the desired performance characteristics for a given jet nebulizer. In an ulysonic nebulizer, the electric energy of alias frequency is used to create vibrational, mechanical energy, typically using a piezoelectric transducer. This energy is transferred to the protein formulation of the central mimetibody composition of the hinge region, EPO mimetic or a specified portion or variant either directly or through a coupling fluid creating an aerosol that includes the the composition of the mimic body of the hinge region, mimetic of the EPO or a specified portion or variant. Advantageously, the particles of the composition of the core composition of the hinge region, mimic of the EPO or a specified portion or variant, supplied by the nebulizer, have a particle size of less than about 10 μm, so preferred, in the range of about 1 μm to about 5 μm, and more preferably about 2 μm to about 3 μm. The formulations of at least one mimic body of the hinge region, EPO mimetic or a specified portion or variant suitable for use with a nebulizer, either jet or ulfrasonic, include, typically, a composition of the composition of the central nervous system. of the hinge region, mimic of the EPO or a specified portion or variant in an aqueous solution at a concentration of about 1 mg to about 20 mg of at least one proiein of the central mimei-body of the hinge region, EPO mimetic or a portion or variant specified per ml of solution. The formulation may include ionic agents such as an excipient, a buffer, an agent for isoyonicity, a preservative, an surfactant and, preferably, zinc. The formulation may also include an excipient or agent for the stabilization of at least one protein of the mimetic composition of the hinge region, mimic of the EPO or a specified portion or variant, as a buffer, a reducing agent., a bulk protein or a carbohydrate. Bulk proteins useful in the formulation of at least one protein of the composition of the central mimetibody of the hinge region, mimetic of EPO or a specified portion or variant include albumin, protamine or the like. Undifferent lipid carbohydrates in the formulation of at least one central membranum of the hinge region, EPO mimic, or a specified portion or variant include sucrose, mannitol, lactose, carbohydrate, glucose, or the like. The at least one central mimetic formulation of the hinge region, EPO mimetic or a specified portion or variant also may include a surfactant, which may reduce or prevent the surface-induced aggregation of at least one central mimetic. of the hinge region, mimetic of EPO or a specified portion or variant, caused by the atomization of the solution to the form an aerosol. Various conventional agents may be employed, such as esters of polyoxyethylene fatty acid and alcohols, and polyoxyethylene sorbital fatty acid esters. The amounts will generally vary between 0.001 and 4% by weight of the formulation. Preferred oxides especially for the purposes of this invention are monooleate polyoxyethylene sorbitan, polysorbafo 80, polysorba 20, or the like. Additional agents known in the art for the formulation of the falic protein as at least one protein of the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant may also be included in the formulation.
Administration of the mimei-body compositions of the hinge region, EPO mimetic or a specified portion or variant by a metered-dose inhaler In a metered-dose inhaler (MDI), a propellant, at least one central mimetic of the region Hinge, mimic of the EPO or a specified portion or variant, and any excipient or other additive, are contained in a box as a mixture including a compressed or liquefied gas. The actuation of the dosing valve releases the mixture as an aerosol, preferably, containing particles in the size range of less than about 10 μm, preferably about 1 μm to about 5 μm, and most preferably about 2 μm to about 3 μm. The desired size of the The aerosol particle can be obtained by employing a formulation of a protein of the composition of the central mimetibody of the hinge region, EPO mimetic or a specified portion or variant, produced by various methods known to those skilled in the art, including jet, spray drying, condensation of the critical point or similar. Preferred metered dose inhalers include those manufactured by 3M or Glaxo and employ a hydrofluorocarbon propellant. Formulations of at least one central mimetibody of the hinge region, EPO mimetic or a portion or variant specified for use with a metered dose inhaler device, will generally include a finely divided powder that contains at least one central mimetic of the region. of hinge, mimetic of the EPO or a portion or variant specified as a suspension in a non-aqueous medium, for example, suspended in a propellant with the aid of an surfactant. The propellant may be any conventional material used for this purpose, such as chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon or a hydrocarbon, including ichlorofluoromethane, dichlorodifluoromethane, dichloroethylfluoroemphenol and 1,1,1- tefrafluoroelan, HFA-134a (hydrofluoroalkane-134a) , HFA-227 (hydrofluoroalkane-227), or similar. Preferably, the propellant is a hydrofluorocarbon. The surfactant can be chosen to stabilize at least one central mimetibody of the hinge region, EPO mimetic or a portion or variant specified as a suspension in the propellant, to protect the active agent from chemical degradation and the like. Suitable lens surfactants include sorbitaniumiumium, soybean lecithin, oleic acid or the like. In some cases, aerosols in solution that use solid solvents such as eneol are preferred. Additional agents known in the art for the formulation of a protein such as a protein can also be included in the formulation. One of ordinary skill in the art will recognize that the methods of the current invention can be achieved by pulmonary administration of at least one composition of a mimic body of the hinge region, mimic of EPO or a portion or variant specified via devices not described. at the moment.
Mucous formulations and administration For the absorption of mucosal surfaces, the compositions and methods for administering at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant, include an emulsion comprising a plurality of submicromeriral particles, a mucoadhesive macromolecule, a bioacid peptide and a phase aqueous coníinua, which promoted the absorption of the mucous surfaces through the mucoadhesion of the particles of the emulsion (U.S. Patent No. 5,514,670). The mucosal surfaces suitable for the application of the emulsions of the present invention they can include routes of administration cornea, conjunctiva, buccal, sublingual, nasal, vaginal, pulmonary, stomach, intestinal and rectal. Formulations for vaginal or rectal administration, for example, suppositories, may contain as excipients, for example, polyalkylene glycols, petrolatum, cocoa butter and the like. Formulations for inanal administration can be solid and contain as excipients, for example, lactose or they can be aqueous or oily solutions of nasal drops. For oral administration, excipients include sugars, calcium esery, magnesium stearate, pregelatinized starch, and the like (U.S. Patent No. 5,849,695).
Oral formulations and administration Formulations for oral administration are based on the co-administration of adjuvants (for example, resorcinols and non-ionic agents, such as polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether), to artificially increase the permeability of intestinal walls. , as well as the co-administration of enzyme inhibitors (eg, pancreatic trypsin inhibitors, diisopropyl fluorophosphate (DFF) and trasilol), to inhibit enzymatic degradation. The active compound of the dosage form of the solid type for oral administration can be mixed with at least one additive, including sucrose, lactose, cellulose, mannitol, euphalosa, raffinose, mallyoyl, dextran, starches, agar, arginases, chitins, chitosan. , pecíinas, gomaragañío rubber, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride. Such dosage forms may also contain other types of additives, for example, an inactive diluent, natural lubricants such as magnesium, paraben, preservatives such as sorbic acid, ascorbic acid, alpha-Icopherol, antioxidants such as cysteine, disinfectants, binders, thickeners, buffer agents, sweeteners, flavoring agents, perfuming agents, etc. Tablets and pills can also be processed in enteric coated preparations. Liquid preparations for oral administration include emulsion, syrup, elixir, suspension and solution preparations, which allow medical use. These preparations may contain inactive diluent agents commonly used in the field, for example, water. Liposomes have also been described as drug delivery systems for insulin and heparin (U.S. Patent No. 4,239,754). More recently, artificial polymer microspheres of mixed amino acids (proteinoids) have been used to deliver the pharmaceuticals (Patenie de E.U.A. No. 4,925,673). In addition, the carrier compounds described in the U.S. Pat. No. 5,879,681 and the U.S. Patent. No. 5,5,871, 753, are used to supply biologically active agents, orally, as is known in the art.
Transdermal Formulations and Administration For transdermal administration, the at least one central mimetibody of the hinge region, EPO mimetic or a specified portion or variant is encapsulated in delivery devices such as liposome or polymer nanoparticles, microcapsule microparticles or microspheres ( collectively referred to as microparticles, unless otherwise indicated). Various 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 orads proteins, alginate and other polysaccharides and combinations thereof (U.S. Patent No. 5,814,599).
Prolonged administration and formulations It may sometimes be desirable to deliver the compounds of the present invention to the subject for extended periods of time, for example, for periods of one week to one year from a single administration. Several dosage forms of release, deposit, or implant can be used. For example, a dosage form may contain a pharmaceutically acceptable non-toxic salt of the compound 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, acid sulfuric acid, cylric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, mono- or disulfonic naphthalenic acid, polygalacryuronic acid and the like; (b) a salt with a polyvalent meifial cation, such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed of for example, N, N ' - dibenzyl-ethylenediamine or eilendiamine; or (c) combinations of (a) and (b), for example, a zinc salt of zinc. Additionally, 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, for example, sesame oil, Suitable for Injection. Particularly preferred salts are zinc salts, zinc salts of zinc, salts of pamoane and the like. Another type of slow release depot formulation for injection would contain the compound or dispersed salt to be encapsulated in a non-toxic, non-antigenic degradation polymer, such as a polylactic acid / polyglycolic acid polymer, for example, as described in the US Patent No. 3,773,919. The compounds or, preferably, the relatively insoluble salts, such as those described above, can also be formulated in cholesterol-matrix syllable granules, particularly for use in animals. In addition, additional slow release, depot or implant formulations, for example, gaseous or liquid liposomes are known in the literature (Pateníe de E.U.A. No. ,770,222 and "Systems of Delivery of Sustained and Concerned Release Drug", J. R. Robinson ed., Marcel Dekker, Inc., N. Y., 1978). Having generally described the invention, it will be more readily understood with reference to the following examples, which are provided by way of illusion and are not intended to be limiting.
EXAMPLE 1 Cloning and expression of a central mimetibody of the hinge region, mimic of EPO in mammalian cells An ichipic mammalian expression vector contains at least one promoter element, which mediates the initiation of transcription of the mRNA, the coding sequence of the mimeiibody of the hinge region, mimetic of EPO or a specified portion or variant, and signals required for the termination of transcription and polyadenylation of the transcript. Additional elements include enhancers, Kozak sequences and nlervinial sequences flanked by donor and acceptor sites for RNA splicing. A highly efficient transcription can be achieved with the early and late promoters for SV40, the long terminal repeats (LTRS) of Reirovirus, for example, RSV, HTLVI, HIVI and the cytomegalovirus early promoter (CMV). However, cell elements can also be used (for example, promoter of human acyin). Expression vectors suitable for use in the practice of the present invention include, for example, vector vectors such as pIRESIneo, pReero-Off, pRetro-On, PLXSN or pLNCX (Clonetech Labs, Palo Alto, CA), pADNc3.1 (+/-), pADNc / Zeo (+ / -) or pADNc3.1 / Hygro (+/-) (Invitrogen), PSVL and PMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109). Mammalian host cells that can be used include Hela 293, H9 and Jurkaf human cells, mouse NIH3T3 and C127 cells, Cos 1 cells, Cos 7 and CV 1, quail QC1-3 cells, mouse and ovarian L cells. of Chinese hamster (CHO). Alternatively, the gene can be expressed in stable cell lines that contain the gene integrated into a chromosome. Co-transfection with a selectable marker such as dhfr, gpf, 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 mimetic core of the hinge region, mimic of the encoded EPO or a specified portion or variant. The DHFR marker (dihydrofolate reductase) is useful for developing cell lines that carry several hundred or even several thousand copies of the gene of interest. Another useful selection marker is the enzyme glutamine synase (GS) (Murphy, et al., Biochem J. 227: 277-279 (1991)).; Bebbingíon, et al., Bio / Technology 10: 169-175 (1992)). Using these markers, the mammalian cells are cultured in a selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified genes integrated in a chromosome. Chinese hamster ovary (CHO) and NSO cells are frequently used for the production of the central mimetibodies of the hinge region, EPO mimetic or a specified portion or variant. The expression vectors pC1 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, for example, with the excision sites of the BamHI resynchronization enzyme, Xbal and Asp718, facilitate the cloning of the gene of interest. The vectors also contain the 3 'infron, the polyadenylation and termination signal of the rabies preproinsulin gene.
Cloning and expression in CHO cells The vector pC4 is used for the expression of the central mimetibody of the hinge region, mimetic of EPO or a specified portion or variant. Plasmid pC4 is a derivative of plasmid pSV2-dhfr (Accession No. ATCC 37146). The plasmid contains the mouse DHFR gene under the conirol of the SV40 early promoter. Chinese hamster ovary cells or cells that lack the activity of dihydrofolate that are transfected with these plasmids can be selected by culturing the cells in a selective medium (eg, alpha minus MEM, Life Technologies, Gaithersburg, MD), supplemented with the meioírexaío quimioferapéuíico agent. The amplification of DHFR genes in methotrexate-resistant (MTX) cells has been well documented (see, for example, F. W. All, al., J. Biol. Chem. 253: 1357-1370 (1978); J. L. Hamlin and C. Ma, Biochem. et Biophys. Acta 1097: 107-143 (1990); and M. J. Page and M. A. Sydenham, Biotechnology 9: 64-68 (1991)). Cells that are grown in concentrations that are increased by MTX develop resistance to the drug by overproducing the target enzyme, DHFR, as a result of the amplification of the DHFR gene. If a second gene binds to the DHFR gene, it is usually co-amplified and overexpressed. It is known in the art that this procedure can be used to develop cell lines that carry more than 1,000 copies of the amplified genes. Subsequently, when the meiofrexa is exhaled, cell lines containing the amplified gene integrated in one or more chromosomes of the host cell are obtained. The plasmid pC4 coniiene to express 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 an isolated fragment of the human cytomegalovirus (CMV) immediate early gene enhancer (Boshart, et al., Cell 41: 521-530 (1985)). Downstream of the promoter is BamHI, Xbal and Asp718, the cleavage sites of the restriction enzyme that allow the integration of genes. Defras of these cloning sites, the plasmid contains the 3 'iniron and the polyadenylation site of the rabies preproinsulin gene. Other promoters of alpha efficiency can also be used for the expression, for example, the human β-actin promoter, the SV40 early or late promoters or the long terminal repeats of other reirovirus, for example, HIV and HTLVI. The expression systems of the Cloneech Tet-Off and Tet-On gene and similar systems can be used to express EPO in a regulated manner in mammalian cells (M. Gossen, and H. Bujard, Proc. Nati. Acad. Sci. USA 89: 5547-5551 (1992)). For the polyadenylation of the mRNA, other signals can also be used, for example, of the human growth hormone or globin genes. Stable cell lines carrying a gene of interest integrated into the chromosomes can also be selected for confraction with selectable markers such as gp1, G418 or hygromycin. It is advantageous to use more than one selectable marker at the start, for example, G418 plus methotrexate. Plasmid pC4 is digested with restriction enzymes and then dephosphorylated using calf intestinal phosphatase by methods known in the art. The vector is then isolated from a 1% agarose gel. The DNA sequence encoding the central mimeinibody of the hinge regionEPO mimic or a specified portion or variant is used, which corresponds to the HC and LC variable regions of a central mimetibody of the hinge region, EPO mimic of the present invention, in accordance with the steps of the method known. The isolated nucleic acid encoding a suitable human conserved region (ie, the HC and LC regions) is also used in this construct. The isolated variable and constant region encoding the DNA and the dephosphorylated vector are then ligated with T4 DNA ligase. The cells of E. coli HB101 or XL-1 Blue are then transformed and the bacteria are identified as containing the fragment inserted in the plasmid pC4 using, for example, analysis of the restriction enzymes. Chinese hamster ovary (CHO) cells lacking an active DHFR gene are used for transfection. 5 μg of the expression plasmid pC4 is contrasted with 0.5 μg of the plasmid pSV2-neo using lipofectin. Plasmid pSV2neo contains a dominant selectable marker, the neo gene of Tn5 that encodes an enzyme that confers resistance to a group of antibiotics, including G418. The cells are seeded in alpha minus MEM supplemented with 1 μg / ml of G418. After 2 days, the cells are fripinized and seeded in hybridoma cloning plates (Greiner, Germany) in alpha minus MEM supplemented with 10, 25 or 50 ng / ml of methotrexate plus 1 μg / ml of G418. After about 10-14 days, the single clones are triptinized and seeded in 6-well Petri dishes or in 10-ml dishes using different methorexame concentrations (50 nM, 100 nM, 200 nM, 400 nM, 800 nM) . The clones that grow at the concentrations more aliquots of meihotrexate are transfered to new plates of 6 wells that condense even greater concentrations of mephoraxate (1 mM, 2 mM, 5 mM, 10 mM, 20 mM). The same procedure was repeated until clones were obtained that grew at a concentration of 100-200 mM. The expression of the prodrug of the desired gene is analyzed, for example, by SDS-PAGE and Western blot or by reverse phase HPLC analysis.
EXAMPLE 2 Non-limiting example of a central mimetibody of the hinge region, mimic of the EPO of the invention Aniecedeníes The EMP-1 (peptide miméíico of the EPO-1) is a peptide of 20 amino acids without homology with the sequence of the human erifropoyeíina (HuEPO), but with the capacity (like a dimer) of acíivar the receiver of the EPO ( Wrighton et al, 1996, Science, vol 273,458-463). However, its relatively low activity (10,000 to 100,000 times less than HuEPO) and its short half-life (ex-vivo half-life of 8 hours in 50% serum, unknown live half-life), compromises its usefulness as a therapeutic agent. Therefore, a way is needed to give the penis a longer half-life, without disturbing, and possibly improving its potency. For this purpose, several attempts have been made to increase the activity of EMP-1 by stabilizing the dimerization of the peptide or by incorporating the peptide into larger structures to increase the half-life. Wrighlen I went to. (1997, Nature Biotechnology, vol.15,1261-65) EMP-1 labeled with biotin combined with streptavidin to isolate dimerization. They observed a 100-fold increase in activity in a cell proliferation assay in vitro. They also used amphi-bioin antibody to stabilize the peptide dimer, however, only a 10-fold increase in activity was observed. The same authors prepared a definite dimeric form chemically of the EMP-1. In this case, a 100-fold increase in in vivo activity was observed. Another group sought to improve the activity of EMP-1 through a covalent bond to polyethylene glycol (PEG) (Johnson et al., 1997, Chem. &Bio., Vol 4 (12), 939-50). They reported an increase in potency up to 1000 times, however, the construct was found to be immunogenic in mice (the antibodies were directed to the peptide) (Dana Johnson, Personal Communications). Kuai I went to. (2000, J. Pepfide Res., Vol 56.59-62) inserted the peptide EMP-1 into the sequence of the plasminogen activator inhibitor-1 (PAI-1). It was thought that the insertion of EMP-1 in this process would make the dimerization and increase the half-life. In an in vivo test, it was observed that the potency of this consfrucio was significantly higher, such as more than 2500 times greater than the EMP-1 alone. It should be noted that different in vilro assays and in vivo models were used in these studies, and the reported potencies may not be comparable with each result or other results presented here.
Central Mimetibody of the Hinge Region, EPO Mimetic of the Present Invention A specific, non-limiting example of this invention is the central EMP-mimic construct of the hinge region, where V is the first of several amino acids N -terminals of a naive HC or LC antibody, P is a single copy of the bioacidive EMP-1 peptide and L is a cascade repeat of the flexible linker Gly-Ser or Gly-Gly-Gly-Ser, H is a region hinge and CH2 and CH3 are the subclasses of the IgG1 or IgG4 isozyme. It is thought that this structure will resist the peptide EMP-1, but will allow sufficient flexibility so that the dimerization of the peptides as part of the assembled homodimer is stabilized. To support this, the EMP-mimetibody activity of the hinge region in an in vitro cell proliferation assay is more than 500 times greater than that of the EMP-1 peptide and only substantially similar to the recombinant HuEPO (rHuEPO). In addition, it is expected that the half-life of this construct will be many times that of the rHuEPO or peptide EMP-1 alone and similar to that of an IgG. Consistently, normal mice treated with the EMP-central mimetibody of the hinge region achieve a maximum hemarocyte significantly more than the mice tested with rHuEPO, when there are units of biological activity, and the elevated levels are maintained. last a longer period. This construct is efficiently secreted from the cells and appears to be folded properly; overcoming the problems associated with the 1st generation mimetibodies. In addition to the basic structure described above, variants with potentially favorable biological characteristics are described. These include those who may have a decreased tendency to self-associate, reduced immune effector functions, or decreased immunogenicity. You will hear modifications that confer the desired characteristics such as the conformation of the biologically active peptide, and the transfer through the blood-brain barrier, they are also considered. The proposed variants and modifications may be combined in any way to provide constructs with the desired activities. Using the recombinant DNA methods, the EMP-1 peptide was inserted into an intermediate vector between an immunoglobulin signal peptide and a human J sequence. This was done using the synthetic oligonucleotides complementary to exíre comparables with the resins of the vector present in the vector. These oligonucleotides comprise coding sequences for the consensus site of the signal peptidase (QIQ), the peptide EMP-1 (SEQ ID NO: 2), and a flexible linker composed of GS or GGGS. A restriction fragment containing the functional elements mentioned above was then transferred to an expression neighbor. This vector contained an ani-CD4 immunoglobulin promoter and enhancer, and the coding sequence for a central sequence of the hinge region of human IgG1, and a portion of a hinge region of IgG1, CPPCP (109- 113 of SEQ ID NO: 66, as shown in Figure 36C), a constant region HC 2 (CH 2) and a consynt region 3 (CH 3), as well as the necessary elements for the replication of the plasmid and the selection in the bacteria and the selection of stable expresores in mammalian cells. This plasmid was linearized and infroduced in the mouse NLE cell melanoma cell line via electroporation. The resistant cells were selected and the high expresores of EMP-mimetibody cenfral of the hinge region, were identified by ELISA assay of culture supernatants. The purification of the cell culture supernatant construct was achieved by affinity chromazography of the standard protein A. The passage of the purified product through polylacrylamide gels containing SDS under denaturation and reduction conditions, confirmed the expected size of the purified product. The identity of the purified protein was further confirmed by mass spectrometry and N terminal sequence. The amino acid sequences of EMP-central amino groups of the hinge region are shown below. Functional domains are indicated above the sequence encoding the peptide. The peptide consensus sequence of the amino acid signal corresponds to the first fresh amino acids of a natural immunoglobulin. It is thought that these amino acids contribute to the efficient removal of the peptide from the signal by the peptidase of the signal in the endoplasmic reticulum. This sequence is immediately followed by the sequence encoding EMP-1. The two C-terminal e-amino acids of the EMP-1 sequence combined with the following six amino acids form a flexible binder characterized by the Gly-Gly-Gly-Ser repeat. A sequence of the human junction region (J) follows later. It is thought that the J sequence will provide even more flexibility to allow the EMP-1 dimer to adopt the proper conformation, and allows the dimer to protrude from the globular structure of the immunoglobulin and penetrate the gap between the two EPO receivers. The hinge region HC is also included in the construct immediately after the region J. There are three cysteines in the hinge region of lgG1 (highlighted). The former would normally mate with the light chain (LC) of the immunoglobulin and the latter two would participate in interchain links between two HCs. The rest of the sequence is composed of the CH2 and CH3 regions, which constitute the volume of the proiein. One of the reasons why immunoglobulins are thought to have a long serum half-life is their ability to bind to FcRn that extends the serum half-life, returning the pinocytosed immunoglobulin back to the ex-cellular space. This FcRn binding site overlaps the junction of the CH2 and CH3 regions (Sheilds et al, 2001, J. Biol. Chem., Vol 276 (9), 6591-6604). The peptide sequence of central EMP-mimetibody of the hinge region shows important functional domains.
V Peptide EMP-1 Hinge Enlanzate lgG1 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GS CPPCP APELLGGP lgG1 CH2 61SVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS lgG1 CH3 122TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAP1EKTISKAKGQPREPQVYTLPPSRDEL lgG1 CH3 183TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ lgG1 CH3 241 QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 82) V Peptide EMP-1 Hinge Enlanzate lgG1 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GGGS CPPCP APELLGGP lgG1 CH2 61 SVFLFPPKPKDTL ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS lgG1 CH3 122TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDEL lgG1 CH3 183TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ lgG1 CH3 241 QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 83) V Peptide EMP-1 Hinge Engage lgG1 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GSGGGS CPPCP APELLGGP lgG1 CH2 61SVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS lgG1 CH3 122TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDEL lgG1 CH3 183TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ lgG1 CH3 241 QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 84) V Peptide EMP-1 Hinge Engage lgG1 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GS CPPCP APEAAGGP lgG1 CH2 61SVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS [gG1 CH3 122TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDEL IgG1 IgG1 CH3 CH3 241 QGNVFSCSVMHEALHNHYTQKSLSLSPGK 183TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ (SEQ ID NO: 85) V EMP-1 Peptide Hinge IgG1 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG Enlanzate GGGS CPPCP APEAAGGP IgG1 CH2 61 SVFLFPP KPKDTLMI SRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS | 9g1 CH3 122TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDEL lgG1 CH3 183TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ lgG1 CH3 241 QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 86) V Peptide EMP-1 Hinge Engage lgG4 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GS CPPCP APEFLGGP lgG4 CH2 61 SVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS lgG4 CH3 121 TYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEM lgG4 CH3 83 TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQ lgG4 CH3 241 EGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 87) V Peptide EMP-1 Hinge Enlanzate lgG4 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GS CPPCP APEAAGGP IgG 4 CH2 61 SVFLFPPKPKDTLMISRTPEVTCWVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS - | gG4 CH3 121 TYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEM lgG4 CH3 183 TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQ lgG4 CH3 241 EGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 88) V Peptide EMP-1 Hinge Enlanzate lgG4 CH2 1 QIQGGTYSCHFGPLTWVCKPQGG GGGS CPPCP APEAAGGP lgG1 CH2 61 SVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS lgG4 CH3 121 TYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEM lgG4 CH3 183 TKNQVSLTCLVKGFYPSDIAVE WESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQ lgG4 CH3 241 EGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 89) It is well known that two heavy chains of IgG are assembled during cellular processing via disulfide bonds between the cysteines located in the hinge region to form a homodimer. This is also expected to occur among the modified peptides to form the assembled construct of EMP-central mimetibody of the hinge region.
In addition, the infra-chain disulfide bond between the two cysteines in the EMP-1 peptide is also expected to be formed. The expected EMP-1 em- emymetry of the hinge region contains two EMP-1 peptides. The spatial arrangement of the N-terminus peptides together with the flexibility of the binding sequences should allow the peptides to form a bioactive dimer. The activity of the EMP mimic-body in the region of the hinge was first tested in an in vitro bioactivity assay. For this test, the EPO-dependent cell line UT-7 / EPO derived from a patient with acute megakaryoblastic leukemia (Komatsu et al., 1993, Blood, vol 82 (2), 456-464) was used. These cells undergo programmed cell death 48 to 72 hours after extraction of the medium supplemented with rHuEPO. Cells that have been incubated in the absence of rHuEPO for 24 hours can be saved if they were brought with rHuEPO or an EPO agonist. The central EMP-mimetibody of the hinge region was added to the cells without food without rHuEPO and the cell viability was determined 48 hours after the treatment using the tetrazolium compound MTS (CelITiter 96 Aque0us One Solution, Promega) which is metabolized by the living cells to provide a production with an absorbance that can be measured. The results of a typical assay showed that the potency of a central EMP-mimetibody of the hinge region on a molar basis is 500 times greater than that of peptide EMP-1 and 5 times less than that of rHuEPO. In addition, these same cells were stimulated with EMP- The central region of the hinge region and the tyrosine phosphorylation pads were visualized by running the cell lysate through a polyacrylamide gel. The pattern exhibited by EMP-mimetic body of the hinge region was similar to that of the rHuEPO, indicating that the mechanism by which the central EMP-mimetibody of the hinge region acts on these cells, is similar to that of the rHuEPO. The in vivo studies were done in normal mice to compare the half-life of the EMP-mimetibody cenfral of the region of the hinge with that of the rHuEPO and to compare its effecíos in the eriíropoyesis. When the mice were dosed equally, the EMP-mimetibody central region of the hinge gave a higher maximum response and the response was prolonged, compared to the rHuEPO. The serum concentrations of the rHuEPO and the central EMP-mimetibody of the hinge region were measured by ELISA. The approximate half-life of the EMP-mlmetic of the hinge region was at least several times that of the rHuEPO. It has been shown that the mutation of two lysine residues (L), L234 and L235, in the lower hinge region of IgG1 to alanine (A), will nullify the immunoglobulin's ability to mediate complement-dependent cyclo-toxicity (CDC) and antibody-dependent cellular toxicity (ADCC) (Hezereh et al., 2001, J. Virol., vol 75 (24), 12161-68). Preliminary stages have shown that the central EMP-mimeiibody of the hinge region does not mediate the complement lysis of the cells expressing the Receiver of the EPO. This may be due to the low number of receptors found in erythroid progenitor cells. In addition, the live expansion of the erythroid progenitors as evidenced by the significant increases in hemaputation supports the possible functional irrelevance of the functions of the immune effector. However, although effects associated with the function of the effector have not been observed, there remains an interest in introducing mutations as a precautionary step. Another modification that would result in a decrease in the mediation of immune effector functions is the elimination of the glycosylation binding site. This can be achieved by the mutation of asparagine at position 297 (N297) to glutamine (Q). Additional changes may optionally include threonine (T) with an alternating amino acid to reduce or modify O-glycosylation, for example, T34 or T47 with Aglucosylated versions of subglass lgG1 known to be poor mediators of effector function immune (Jefferis et al., 1998, Immol., Rev., 163, 50-76).
Advantages The novel construct, EMP-mimetibody central region of the hinge described above offers an alternate way to show the bioactive peptide EMP-1. The activity of this construct is in the range of the rHuEPO and the average life is similar to that of an IgG.
In addition, it is expected that the proposed modifications, in combination and with the addition to the novel characteristics of the EMP-mimetibody cenfral of the hinge region, improve the uty of the construct of EMP-central mimetibody of the region of the hinge. It is clear that the invention can be practiced in a manner as described particularly in the description and the examples above. Numerous modifications and variations of the present invention are possible, in light of the foregoing teachings and, as a result, are beyond the scope of the present invention.

Claims (40)

NOVELTY OF THE INVENTION CLAIMS
1. A nucleic acid of a mimic body of the hinge region, mimic of EPO, comprising at least one polynucleotide encoding at least one amino acid sequence of SEQ ID NOS: 82 and 84, or a complementary polynucleotide thereof.
2. A nucleic acid of a central mimetibody of the hinge region, mimic of EPO, comprising at least one polynucleotide encoding at least one amino acid sequence of SEQ ID NOS: 83 and 85-89, or a polynucleotide complementary to it.
3. A nucleic acid of a central mimetibody of the hinge region, mimic of EPO, comprising at least one polynucleotide encoding at least one amino acid sequence of SEQ ID NOS: 1-30, or a polynucleotide complementary to the same.
4. A nucleic acid of a central mimetibody of the hinge region, mimic of EPO, comprising at least one polynucleotide encoding a polypeptide according to Formula (I): ((V (m) -P (n ) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of an N term of an immunoglobulin variable region, P is at least one mimetic mimic of bioactive EPO, L is a linking sequence, H is at least a portion of a central variable region of the immunoglobulin hinge, 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, rys can independently be an integer 0, 1 or 2 and 10.
5. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising at least all contiguous amino acids of at least one of SEQ ID NO: 82 and 84.
6.- A polypeptide of a central mimetibody of the hinge region, EPO mimetic, comprising at least all of the contiguous amino acids of at least one of SEQ ID NO: 83 and 85-89.
7. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising at least all contiguous amino acids of at least one of SEQ ID NOS: 1-30.
8. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) - H (p) -CH2 (q) -CH3 (r)) (s), wherein V is QIQ, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L comprises GS, GGGS (SEQ ID NO: 73) or GSGGGS (SEQ ID NO: 74), H is CPPCP (SEQ ID NO: 75), CH2 is APELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGV EVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK TISKAK (SEQ ID NO: 76), CH3 is GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 78), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
9.- A polypeptide of a central mimei-body of the region hinge, EPO mimetic, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 ( r)) (s), wherein V is QIQ, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L comprises GS, GGGS (SEQ ID NO: 73) or GSGGGS (SEQ ID NO: 74), H is CPPCP (SEQ ID NO: 75), CH2 is APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDG VEVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAK (SEQ ID NO: 77), CH3 is GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 78), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
10.- A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is QIQ, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L comprises GS, GGGS (SEQ ID NO: 73) or GSGGGS (SEQ ID NO. : 74), H is CPPCP (SEQ ID NO: 75), CH2 is APEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDG EVHNAKTKPREEQFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEK TISKAK (SEQ ID NO: 79), CH3 is GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 81), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
11.- A polypeptide of a mimeiibody central to the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is QIQ, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L comprises GS, GGGS (SEQ ID NO: 73) or GSGGGS (SEQ ID NO. : 74), H is CPPCP (SEQ ID NO: 75), CH2 is APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDG VEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIE KTISKAK (SEQ ID NO: 80), CH3 is GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 81), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
12.- A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is a N-terminal portion of a human variable region, P is at least one peptide bioacid selected from SEQ ID NOS: 1-30, L is a binding polypeptide, H is at least a portion of a variable region of the immunoglobulin hinge, CH2 is APELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGV EVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK TISKAK (SEQ ID NO: 76), CH3 is GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 78), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
13.- A polypeptide of a mimetic body of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is a N-terminal portion of a human variable region, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L is a linker polypeptide, H is at least a portion of a central variable region of the immunoglobulin hinge, CH2 is APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDG VEVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAK (SEQ ID NO: 77), CH3 is GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 78), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
14. A polypeptide of a mimeiibody of the region of hinge, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r) )) (s), wherein V is a N-terminal portion of a human variable region, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L is a linker polypeptide, H is at least one portion of a central variable region of the immunoglobulin hinge, CH2 s APEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDG VEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIE KTISKAK (SEQ ID NO: 79), CH3 is GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 81), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
15.- A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is a N-terminal portion of a human variable region, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L is a linker polypeptide, H is at least a portion of a central variable region of the immunoglobulin hinge, CH2 is APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNWYVDG VEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIE KTISKAK (SEQ ID NO: 80), CH3 is GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 81), and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
16.- A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is QIQ, P is at least one bioacid peptide selected from SEQ ID NOS: 1-30, L is a linker polypeptide, H is at least a portion of a central variable region of the immunoglobulin hinge, CH2 is at least a portion of a CH2 immunoglobulin constant region, CH3 is at least a portion of a CH3 immunoglobulin constant region, and m, n, o, p, q, r, s are independently an integer between 0, or 2 and 10.
17. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising an acupressure polypeptide. with the Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of a N term of a variable immunoglobulin region, P is at least a bioactive EPO mimic peptide, L comprises GS, GGGS (SEQ ID NO: 73) or GSGGGS (SEQ ID NO: 74), H is at least a portion of a variable region of the immunoglobulin hinge, CH2 is at minus one portion of a region consists of CH2 of immunoglobulin, CH3 is at least a portion of a CH3 constant region of immunoglobulin, and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
18. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of an N terminus of an immunoglobulin variable region, P is at least one peptide mimetic of Bioacid EPO, L is a binding polypeptide, H is at least a portion of a variable region of the immunoglobulin hinge, CH2 is at least a portion of an immunoglobulin CH2 constant region, CH3 is at least a portion of a CH3 region of immunoglobulin, and m, n, o, p, q, r, s are independently an integer enire 0, 1 or 2 and 10.
19.- A polypeptide of a central mimetibody l of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H (p) -CH2 (q ) -CH3 (r)) (s), wherein V is at least a portion of an N terminus of a variable immunoglobulin region, P is at least a bioactive EPO mimic peptide, L is a binding polypeptide, H is CPPCP (SEQ ID NO: 75), CH2 is at least a portion of a region consisting of CH2 of immunoglobulin, CH3 is at least a portion of a CH3 constant region of immunoglobulin, and m, n, o, p, q, r, s are independently an integer between 0, 1 or 2 and 10.
20. A polypeptide of a central mimetibody of the hinge region, mimic of EPO, comprising a polypeptide according to Formula (I): ((V (m) -P (n) -L (o) -H ( p) -CH2 (q) -CH3 (r)) (s), wherein V is at least a portion of an N term of an immunoglobulin variable region, P is at least one bioactive peptide selected from SEQ ID NOS: 1-30, L is a binding polypeptide, H is at least a portion of a central variable region of the immunoglobulin hinge, CH2 is at least a portion of a CH2 immunoglobulin constant region, CH3 is at least a portion of a region consisting of immunoglobulin CH3, and m, n, o, p, q, r, s are independently an integer enire 0, 1 or 2 and 10.
21.- A nucleic acid of a central mimetibody of the hinge region, mimic of EPO or a polypeptide of a central mimetibody of the hinge region, EPO mimic, according to at least one of claims 1-20, wherein the polypeptide Eptide has at least one activity of at least one P polypeptide.
22. A monoclonal or polyclonal anti-idiotype antibody, fusion protein or fragment thereof, which specifically binds to at least one polypeptide of a central mimetibody of the hinge region. , EPO mimetic, according to at least one of claims 5-20.
23. A nucleic acid of a central mimetibody of the hinge region, mimic of EPO, which encodes at least one polypeptide of a mimic body of the hinge region, mimic of EPO or a antibody of a central mimetibody of the hinge region, mimic of EPO, according to any of claims 1-20.
24. A vector of a mimeiibody of the hinge region, mimic of EPO, comprising at least one isolated nucleic acid according to claim 23.
25.- A host cell of a mimic body of the hinge region , EPO mimic, comprising an isolated nucleic acid according to claim 23.
26.- The host cell of a central mimetibody of the hinge region, mimic of the EPO, according to claim 23, further characterized because the host cell is at least one selected from COS-1, COS-7, HEK293, BHK21, CHO, BSC-1, Hep G2, 653, SP2 / 0, 293, NSO, DG44 CHO, CHO K1, HeLa, of myeloma or lymphoma, or any cell derived, immortalized or transformed from them.
27. A method for producing at least one polypeptide of a central mimetibody of the hinge region, mimetic of EPO or an antibody of a mimetic body of the hinge region, mimic of EPO, which comprises translating a nucleic acid of according to claim 23 under in vitro conditions, either in vivo or in situ, so that the central mimetibody of the hinge region, mimetic of the EPO or the antibody is expressed in detectable or recoverable amounts.
28. A composition comprising at least one nucleic acid of a central mimetibody of the hinge region, mimic of EPO, a polypeptide of a mimei-body of the hinge region, mimetic of EPO, or an antibody of a central antibody mimic. the hinge region, mimic of EPO, according to at least one of claims 1-20.
29. The composition according to claim 28, further characterized in that the composition further comprises at least one pharmaceutically acceptable carrier or diluent.
30. The composition according to claim 28, further characterized in that it comprises at least one composition comprising a therapeutically effective amount of at least one compound, composition or polypeptide selected from at least one of a deicible brand or reporter, an antagonist of the TNF, an anti-infective drug, a drug for the cardiovascular system (CV), a drug for the central nervous system (CNS), a drug for the autonomic nervous system (ANS), a drug for the respiratory tract, a drug for the tract Gastrointestinal (Gl), a hormonal drug, a fluid or electrolyte balance drug, a hematologic drug, an antineoplastic, an immunomodulation drug, an ophthalmic, otic or nasal drug, a topical drug, a nutritional drug, a cytokine or a cytokine antagonist.
31. The composition according to claim 28, further characterized in that it is in a form of at least one selected from a liquid, gas or solution, mixture, suspension, emulsion or dry colloid, a lyophilized preparation or a powder.
32.- The use of at least one nucleic acid, polypeptide or antibody of a mimetic body of the hinge region, mimic of the EPO, according to at least one of claims 1-20, for preparing a composition for diagnosing or treating a condition related to the EPO ligand in a cell, tissue, organ or animal.
33. The use claimed in claim 32, wherein the composition comprises 0.001-50 mg of the antibody of the mimetibody cenfral of the hinge region, mimic of EPO; 0.000001-500 mg of the mimetic body of the hinge region, EPO mimetic; or 0.0001-100 μg of the nucleic acid of the central mimetibody of the hinge region, EPO mimic per kilogram of cells, tissue, organ or animal.
34. The use claimed in claim 32, wherein the composition is formulated to be administrable by at least one selected parenteral mode, subcutaneous, iníramuscular, infravenous, infraarticular, inírabronchial, iníraabdominal, iníracapsular, infracartilaginous, inirachial, inraceal, intracelebelar, intracerebroventricular, intracholic, intracervical, intragastric, intrahepatic, intramyocardial, intraosy, inírapélvico, nírapericárdico, iníraperifoneal, infrapleural, iníraprosíático, intrapulmonar, Intrarectal, intrarenal, nireretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, intralesional, bolus, vaginal, recial, buccal, sublingual, iníranasal or fransdermal.
The use claimed in claim 32, wherein said composition is administerable before, concurrently or subsequently, at least one composition comprising an effective amount of at least one compound or polypeptide selected from at least one of a Defective brand or reporter, a TNF antagonist, an antiepileptic drug, a drug for the cardiovascular system (CV), a drug for the central nervous system (CNS), a drug for the autonomic nervous system (ANS), a drug for the respiratory tract, a drug for the gastrointestinal tract (Gl), a hormonal drug, a fluid balance or electrolyte drug, a blood drug, an antineoplastic, a drug for immunomodulation, an ophthalmic, otic or nasal drug, a topical drug, a nufritivo drug, a cytokine or a cifocin antagonist.
36.- A device comprising at least one polypeptide, antibody or nucleic acid of an isolated central mimetibody of the mimetic hinge region of the EPO, according to at least one of claims 1-20, wherein the device is suitable to bring into contact or administer at least one of the polypeptide, antibody or nucleic acid of a central mimetibody of the hinge region, EPO mimetic, mediating at least one selected mode of parenteral, subcutaneous, intramuscular, intravenous, intraarticular, intrabronchial , Intraabdominal, incacapsular, incarartilaginous, intracavity, intracellular, ntracelebelar, intracerebroventricular, intracranial, intracervical, iníragásirico, inirahepalic, inréchalcardic, iníraósieo, nellarílco, nírapericardico, inraperiíoneal, nfrapleural, infraprosíático, intrapulmonar, inírarrectal, intrarenal, intraretinal, intratraspinal, intrasinovial, intraforácico, intrauterine, intravesical, iníralesional, bolo, vaginal, recial, buccal, sublingual, iníranasal or transdermal.
37. An article of manufacture for pharmaceutical use or diagnosis in humans, comprising a packaging material and a container comprises at least one polypeptide, anibody or nucleic acid of a central isolated mimetibody of the mimic hinge region of the EPO, according to at least one of claims 1-20.
38.- The article of manufacture according to claim 37, further characterized in that the container is a component of a parenteral, subcutaneous, intramuscular, intravenous, narticular, inirabronchial, intraabdominal, intracapsular, intracartilage, intracavity, or parenteral device or delivery system. intracelial, intracelebelar, ntracerebroventricular, intracolic, níracervical, níragásírico, inírahepáíico, iníramiocárdico, intraósteo, inírapélvico, intrapericardial, intraperitoneal, intrapleural, intraprosíáíico, intrapulmonary, intrarrecíal, inírarrenal, infrarreíinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, intralesional , bolus, vaginal, rectal, buccal, sublingual, intranasal or transdermal.
39. A method for producing at least one polypeptide, antibody or nucleic acid of a mimei-body isolated from the mimic hinge region of the EPO, in accordance with at least one of the claims 1-20, comprising providing at least one host cell, transgenic animal, transgenic plant, plant cell, capable of expressing in delectable or recoverable amounts the polypeptide, antibody or nucleic acid. 40.- At least one polypeptide, antibody or nucleic acid of a mimic member of the hinge region, mimic of the EPO, produced by a method in accordance with claim 39.
MXPA/A/2006/003677A 2003-09-30 2006-03-30 Human epo mimetic hinge core mimetibodies, compositions, methods and uses MXPA06003677A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/507,349 2003-09-30

Publications (1)

Publication Number Publication Date
MXPA06003677A true MXPA06003677A (en) 2006-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
US7241733B2 (en) Mammalian EPO mimetic CH1 deleted mimetibodies, compositions, methods and uses
AU2004277884B2 (en) Human EPO mimetic hinge core mimetibodies, compositions, methods and uses
US7718176B2 (en) Human EPO mimetic hinge core mimetibodies, compositions, methods and uses
US8071103B2 (en) Pharmaceutical composition comprising a human GLP-1 mimetibody
WO2004002417A2 (en) Mammalian ch1 deleted mimetibodies, compositions, methods and uses
WO2005081687A2 (en) Human hinge core mimetibodies, compositions, methods and uses
JP2007508011A (en) Human hinge core mimetibody, compositions, methods and uses
MXPA06011425A (en) Human glp-1 mimetibodies, compositions, methods and uses.
MXPA06003677A (en) Human epo mimetic hinge core mimetibodies, compositions, methods and uses
AU2011202563A1 (en) Human EPO mimetic hinge core mimetibodies, compositions, methods and uses