US20190134196A1 - Etar antibody, and pharmaceutical compositions and use thereof - Google Patents

Etar antibody, and pharmaceutical compositions and use thereof Download PDF

Info

Publication number
US20190134196A1
US20190134196A1 US16/305,828 US201716305828A US2019134196A1 US 20190134196 A1 US20190134196 A1 US 20190134196A1 US 201716305828 A US201716305828 A US 201716305828A US 2019134196 A1 US2019134196 A1 US 2019134196A1
Authority
US
United States
Prior art keywords
seq
formulation
amino acid
concentration
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/305,828
Other languages
English (en)
Inventor
Cheng Zhang
Kesuo Fan
Yong Guo
Chenjiang YAO
Hua Zhang
Xiaofeng Wang
Shuqian Jing
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gmax Biopharm LLC
Original Assignee
Gmax Biopharm LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CN201610954533.2A external-priority patent/CN107987162A/zh
Application filed by Gmax Biopharm LLC filed Critical Gmax Biopharm LLC
Publication of US20190134196A1 publication Critical patent/US20190134196A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2869Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against hormone receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • an ET A R antibody and a pharmaceutical composition thereof for example, a stable pharmaceutical solution formulation of the ET A R antibody. Also provided herein is a method of treating, preventing, or alleviating one or more symptoms of pulmonary arterial hypertension or one or more symptoms of cancer of a reproductive organ.
  • ET Endothelin
  • ET is a vasoconstriction peptide hormone, important to the homeostasis and regulation of the biological functions of the cardiovascular system. ET is found not only in the endothelium but also in many other tissues and cell types (Barton et al., 2008 , Can. J. Physiol. Pharmacol. 86:485-498). ET is a 2,400 Da peptide of 21 amino acids, having 2 disulfide bonds at its N-terminus, linking the 1st and 15th cysteine residues and the 3rd and 11th cysteine residues, respectively. Its C-terminus contains hydrophobic amino acid residues.
  • ET-1 N-terminal structure is important for binding to its receptor, while its C-terminal structure is important as to where on the receptor to bind.
  • ET has three isoforms: ET-1, ET-2 and ET-3. They differ by a few amino acid residues. ET-1 plays a major role in the regulation of the biological functions of the cardiovascular system. Upon stimulation, endothelial cells synthesize and release ET-1. ET-1 is mainly regulated at the transcription level.
  • ET A R Endothelin receptors
  • ETBR Endothelin receptors
  • GPCR G protein-coupled receptor
  • ET A R activates membrane Na + /Ca 2+ exchanger (NCX) and Na + /H + exchanger (NHE) to increase cellular Ca 2+ concentrations and to sensitize muscle fibers to Ca 2+ , resulting in the constriction of vascular smooth muscle and cardiac muscle (Neylon, 1999 , Clin. Exp. Pharmacol. Physiol. 26:149-153).
  • ETBR mainly relaxes the vascular smooth muscle cells and cardiac muscle cells (Nelson et al., 2003 , Nat. Rev. Cancer 3:110-113).
  • ET A R belongs to GPCR family A and has seven transmembrane domains.
  • the extracellular domain is short and small, and only accounts for about one seventh of the full-length receptor, while a GPCR antibody can only target its extracellular domain.
  • Pulmonary arterial hypertension is due to the vasoconstriction of the lung or lung related vasculature, resulting in lung artery insufficiency and a compensatory increase in the blood pressure of the heart.
  • PAH Pulmonary arterial hypertension
  • PAH is a disease with a fairly high rate of disability or death. It is a devastating disease that severely affects the health of patients and imposes significant burden on society.
  • PAH The severity of PAH depends on the degree of relevant cardiac deformity, and the common congenital cardiac abnormalities that will result in secondary PAH includes: aortic stenosis, aortopulmonary window, atrial septal defect, complete atrioventricular septal defect, artery coarctation, dilated cardiomyopathy, double outlet right ventricle, hypertrophic cardiomyopathy, mitral stenosis, patent ductus arteriosus, single ventricle, persistent truncus arteriosus, and ventricular septal defect.
  • PAH mainly affects pulmonary arteries and right heart, causing right ventricular hypertrophy, right atrial dilatation, the dilatation of the trunk of the pulmonary artery, and the sparsity of the surrounding pulmonary arterioles.
  • the hypertrophy of endothelial and smooth muscle cells of pulmonary arteriole results in tunica intima fibrosis, tunica media hypertrophy, lumina stenosis, occlusion or distortion, and plexus change. Tunica intima fibrosis and lumina occlusion may also afflict the pulmonary venules.
  • an ET A R antagonist can effectively block the increase in vascular pressure caused by endothelin to ameliorate PAH symptoms and improve exercise capability and hemodynamics in PAH patients (Serasli et al., 2010 , Recent Pat. Cardiovasc. Drug Discov. 5:184-95).
  • the antibody provided herein can specifically bind to a human ET A R and attenuate pulmonary arterial pressure in an animal model. It can significantly improve a symptom of PAH in an animal model.
  • an ET A R antibody and a pharmaceutical composition thereof for example, a stable pharmaceutical solution formulation of the ET A R antibody. Also provided herein is a method of treating, preventing, or alleviating one or more symptoms of pulmonary arterial hypertension or one or more symptoms of cancer of a reproductive organ.
  • the ET A R antibody provided herein comprises 1, 2, 3, 4, 5, or 6 amino acid sequences, wherein each amino acid sequence is independently selected from:
  • composition comprising an ET A R antibody provided herein and one or more pharmaceutically acceptable carriers.
  • a stable pharmaceutical solution formulation of an ET A R antibody comprising an ET A R antibody provided herein and a buffer.
  • a method of treating, preventing or alleviating one or more symptoms of pulmonary arterial hypertension in a subject comprising administrating to the subject a therapeutically effective amount of a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • a method of treating, preventing, or alleviating one or more symptoms of a disease associated with elevated pulmonary arterial pressure in a subject comprising administrating to the subject a therapeutically effective amount of a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • a method of treating, preventing, or alleviating one or more symptoms of cancer of a reproductive organ in a subject comprising administrating to the subject a therapeutically effective amount of a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • kits for treating pulmonary arterial hypertension, a disease associated with elevated pulmonary arterial pressure, or cancer of a reproductive organ comprising a pharmaceutical composition provided herein.
  • a pharmaceutical composition provided herein in the manufacture of a medicament for treating pulmonary arterial hypertension, a disease associated with elevated pulmonary arterial pressure, or cancer of a reproductive organ.
  • nucleic acid comprising a polynucleotide sequence encoding an ET A R antibody provided herein.
  • a recombinant expression vector comprising a nucleic acid provided herein.
  • a host cell comprising a vector provided herein.
  • Provided herein is a method for producing an ET A R antibody, comprising cultivating a host cell under conditions suitable for expressing an antibody provided herein.
  • FIG. 1 shows the ELISA screening results of the supernatants of hybridomas for binding to CHO-DHFR-ET A R cells (labeled as CHO-ET A R in the figure).
  • the ET A R antibody A-1 (comprising SEQ ID NO: 138 and SEQ ID NO: 166) was obtained from hybridoma clone 15F3.
  • FIG. 2 shows the specific binding of recombinant ET A R antibodies (A-1, A-2 (comprising SEQ ID NO: 140 and SEQ ID NO: 168), A-7 (comprising SEQ ID NO: 150 and SEQ ID NO: 178), and A-12 (comprising SEQ ID NO: 160 and SEQ ID NO: 188)) to human ET A R as determined by FACS.
  • the gray peak and the dotted peak are negative controls, the dotted peak representing the binding curve of the ET A R antibody to CHO-DHFR- and the solid line peak representing the binding curve of the ET A R antibody to CHO-DHFR-ET A R.
  • FIG. 3 shows the inhibitory effects of the supernatants of hybridomas on cellular ET A R-mediated Ca 2+ changes as determined using a calcium flux assay.
  • FIG. 5 shows the in vivo activity of the recombinant ET A R (A-1) in a hypoxia-induced PAH cynomolgus monkey model.
  • A-1 was found to be able to reduce the hypoxia-induced pulmonary systolic pressure significantly, and also to be effective within 96-hr as measured by area under the curve of the pulmonary systolic pressure versus time.
  • FIG. 6 shows that the biological activity of ET A R antibody A-1 (25 mg/mL) did not change significantly after 3 months of storage in a formulation solution containing 20 mM sodium citrate, 140 mM arginine-HCl, and 0.04% TWEEN-80 at pH 5.8 and 4° C.
  • FIG. 7 shows that the biological activity of ET A R antibody A-1 (25 mg/mL) did not change significantly after 3 months of storage in a formulation solution containing 20 mM sodium citrate, 140 mM arginine-HCl, and 0.04% TWEEN-80 at pH 5.8 and 25° C.
  • polypeptide sequences have their amino termini at the left and their carboxyl termini at the right, and single-stranded nucleic acid sequences and the top strands of double-stranded nucleic acid sequences have their 5′ termini at the left and their 3′ termini at the right.
  • a particular section of a polypeptide can be designated by amino acid residue numbers such as amino acids 80 to 130, or in combination with the corresponding actual residues such as Lys80 to Lys130.
  • a particular polypeptide or polynucleotide sequence also can be described by showing its differences from a reference sequence.
  • peptide “polypeptide” and “protein” each refer to a molecule comprising two or more amino acid residues joined to each other by peptide bonds. These terms encompass, e.g., native and artificial proteins, protein fragments and polypeptide analogs (such as muteins, variants, and fusion proteins) of a protein sequence as well as post translationally, or otherwise covalently or non-covalently, modified proteins.
  • a peptide, polypeptide, or protein may be monomeric or polymeric.
  • polypeptide fragment refers to a polypeptide that has an amino-terminal and/or carboxyl-terminal deletion as compared to a corresponding full-length protein. Fragments can be, for example, at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 50, 70, 80, 90, 100, 150 or 200 amino acids in length. Fragments can also be, for example, at most 1,000, 750, 500, 250, 200, 175, 150, 125, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 14, 13, 12, 11, or 10 amino acids in length.
  • a fragment can further comprise, at either or both of its ends, one or more additional amino acids, for example, a sequence of amino acids from a different naturally-occurring protein (e.g., an Fc or leucine zipper domain) or an artificial amino acid sequence (e.g., an artificial linker sequence).
  • additional amino acids for example, a sequence of amino acids from a different naturally-occurring protein (e.g., an Fc or leucine zipper domain) or an artificial amino acid sequence (e.g., an artificial linker sequence).
  • Polypeptides of the disclosure include polypeptides that have been modified in any way and for any reason, for example, to: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter susceptibility to form a protein complex, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties.
  • Analogs include muteins of a polypeptide. For example, single or multiple amino acid substitutions (e.g., conservative amino acid substitutions) can be made in the naturally occurring sequence (e.g., in the portion of the polypeptide outside the domain(s) forming intermolecular contacts).
  • a “conservative amino acid substitution” is one that does not substantially change the structural characteristics of the parent sequence (e.g., replacement of an amino acid should not break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterize the parent sequence or are necessary for its functionality).
  • a “variant” of a polypeptide comprises an amino acid sequence wherein one or more amino acid residues are inserted into, deleted from and/or substituted into the amino acid sequence relative to another polypeptide sequence.
  • Variants of the disclosure include fusion proteins.
  • a “derivative” of a polypeptide is a polypeptide that has been chemically modified, e.g., via conjugation to another chemical moiety such as, for example, polyethylene glycol, albumin (e.g., human serum albumin), phosphorylation, and glycosylation.
  • another chemical moiety such as, for example, polyethylene glycol, albumin (e.g., human serum albumin), phosphorylation, and glycosylation.
  • antibody includes, in addition to antibodies comprising two full-length heavy chains and two full-length light chains, derivatives, variants, fragments, and muteins thereof, examples of which are described below.
  • an “antibody” is a protein comprising a portion that binds to an antigen and optionally a scaffold or framework portion that allows the antibody to adopt a conformation that promotes the binding of the antibody to the antigen.
  • antibodies include antibodies, antibody fragments (e.g., an antigen binding portion of an antibody), antibody derivatives, and antibody analogs.
  • the antibody can comprise, for example, an alternative protein scaffold or artificial scaffold with grafted CDRs or CDRs derivatives.
  • Such scaffolds include, but are not limited to, antibody-derived scaffolds comprising mutations introduced, for example, to stabilize the three-dimensional structure of the antibody as well as completely synthetic scaffolds comprising, for example, a biocompatible polymer.
  • PAMs peptide antibody mimetics
  • An antibody can have, for example, the structure of a naturally occurring immunoglobulin.
  • An “immunoglobulin” is a tetrameric molecule. In a naturally occurring immunoglobulin, each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxyl terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as kappa and lambda light chains.
  • Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids.
  • the heavy chain also includes a “D” region of about 10 more amino acids. See generally, Fundamental Immunology, Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)) (incorporated herein by reference in its entirety for all purposes).
  • the variable regions of each light/heavy chain pair form the antibody binding site such that an intact immunoglobulin has two binding sites.
  • Naturally occurring immunoglobulin chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. From N-terminus to C-terminus, both light and heavy chains comprise regions FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each region is in accordance with the definitions of Kabat et al. in Sequences of Proteins of Immunological Interest, 5th Ed., US Dept. of Health and Human Services, PHS, NIH, NIH Publication No. 91-3242, 1991.
  • an “antibody” refers to an intact immunoglobulin or to an antigen binding portion thereof that competes with the intact antibody for specific binding.
  • Antigen binding portions can be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies.
  • Antigen binding portions include, inter alia, Fab, Fab′, F(ab′) 2 , Fv, domain antibodies (dAbs), fragments including complementarity determining regions (CDRs), single-chain antibodies (scFv), chimeric antibodies, diabodies, tribodies, tetrabodies, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.
  • a Fab fragment is a monovalent fragment having the V L , V H , C L and C H1 regions; a F(ab′) 2 fragment is a bivalent fragment having two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment has the V H and C H1 regions; and a dAb fragment has a V H region, a V L region, or an antigen-binding fragment of a V H or V L region (U.S. Pat. Nos. 6,846,634, 6,696,245, US App. Pub. Nos. 05/0202512, 04/0202995, 04/0038291, 04/0009507, 03/0039958, Ward et al., 1989 , Nature 341:544-546).
  • a single-chain antibody is an antibody in which a V L and a V H region are joined via a linker (e.g., a synthetic sequence of amino acid residues) to form a continuous protein chain wherein the linker is long enough to allow the protein chain to fold back on itself and form a monovalent antigen binding site (see, e.g., Bird et al., 1988 , Science 242:423-26 and Huston et al., 1988 , Proc. Natl. Acad. Sci. USA 85:5879-83).
  • a linker e.g., a synthetic sequence of amino acid residues
  • Diabodies are bivalent antibodies comprising two polypeptide chains, wherein each polypeptide chain comprises V H and V L regions joined by a linker that is too short to allow for pairing between two regions on the same chain, thus allowing each region to pair with a complementary region on another polypeptide chain (see, e.g., Holliger et al., 1993 , Proc. Natl. Acad. Sci. USA 90:6444-48, and Poljak et al., 1994 , Structure 2:1121-23). If the two polypeptide chains of a diabody are identical, then a diabody resulting from their pairing will have two identical antigen binding sites. Polypeptide chains having different sequences can be used to make a diabody with two different antigen binding sites. Similarly, tribodies and tetrabodies are antibodies comprising three and four polypeptide chains, respectively, and forming three and four antigen binding sites, respectively, which can be the same or different.
  • Complementarity determining regions (CDRs) and frame work regions (FR) of a given antibody can be identified using the system described by Kabat et al. in Sequences of Proteins of Immunological Interest, 5th Ed., US Dept. of Health and Human Services, PHS, NIH, NIH Publication no. 91-3242, 1991.
  • One or more CDRs can be incorporated into a molecule either covalently or noncovalently to make it an antibody.
  • An antibody can incorporate the CDR(s) as part of a larger polypeptide chain, can covalently link the CDR(s) to another polypeptide chain, or can incorporate the CDR(s) noncovalently.
  • the CDRs permit the antibody to specifically bind to a particular antigen of interest.
  • An antibody can have one or more binding sites. If there are more than one binding sites, the binding sites can be identical to one another or can be different. For example, a naturally occurring human immunoglobulin typically has two identical binding sites, while a “bispecific” or “bifunctional” antibody has two different binding sites.
  • murine antibody includes all antibodies that have one or more variable and constant regions derived from a murine immunoglobulin sequence.
  • humanized antibody refers to an antibody that produced by grafting the complementarity determining region sequence of a murine antibody molecule into a human antibody variable region framework.
  • antibody-binding site is a portion of an antibody that comprises amino acid residues (or other portion) interacting with an antigen and contributing to the specificity and affinity of the antibody for the antigen. For antibodies that specifically bind to their antigen, this will include at least a portion of at least one of its CDR regions.
  • epitope is the portion of a molecule that is bound by an antibody (e.g., by an antibody).
  • An epitope can comprise non-contiguous portions of the molecule (e.g., in a polypeptide, amino acid residues that are not contiguous in the polypeptide's primary sequence but that, in the context of the polypeptide's tertiary and quaternary structure, are near enough to each other to be bound by an antibody).
  • the “percent identity” of two polynucleotide or two polypeptide sequences is determined by comparing the sequences using the GAP computer program (a part of the GCG Wisconsin Package, version 10.3 (Accelrys, San Diego, Calif.)) using its default parameters.
  • nucleic acid molecules e.g., cDNA or genomic DNA
  • RNA molecules e.g., mRNA
  • analogs of the DNA or RNA generated using nucleotide analogs e.g., peptide nucleic acids and non-naturally occurring nucleotide analogs
  • hybrids thereof e.g., peptide nucleic acids and non-naturally occurring nucleotide analogs
  • the nucleic acid molecule can be single-stranded or double-stranded.
  • the nucleic acid molecules of the invention comprise a contiguous open reading frame encoding an antibody of the invention, or a fragment, derivative, mutein, or variant thereof.
  • Two single-stranded polynucleotides are “the complement” of each other if their sequences can be aligned in an anti-parallel orientation such that every nucleotide in one polynucleotide is opposite its complementary nucleotide in the other polynucleotide, without the introduction of gaps and without unpaired nucleotides at the 5′ or the 3′ end of either sequences.
  • a polynucleotide is “complementary” to another polynucleotide if the two polynucleotides can hybridize to one another under moderately stringent conditions.
  • a polynucleotide can be complementary to another polynucleotide without being its complement.
  • vector is a nucleic acid that can be used to introduce another nucleic acid linked to it into a cell.
  • a vector refers to a linear or circular double stranded DNA molecule into which additional nucleic acid segments can be ligated.
  • a viral vector e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses
  • certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors comprising a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • An “expression vector” is a type of vectors that can direct the expression of a chosen polynucleotide.
  • a nucleotide sequence is “operably linked” to a regulatory sequence if the regulatory sequence affects the expression (e.g., the level, timing, or location of expression) of the nucleotide sequence.
  • a “regulatory sequence” is a nucleic acid that affects the expression (e.g., the level, timing, or location of expression) of a nucleic acid to which it is operably linked.
  • the regulatory sequence can, for example, exert its effects directly on the regulated nucleic acid, or through the action of one or more other molecules (e.g., polypeptides that bind to the regulatory sequence and/or the nucleic acid).
  • Examples of regulatory sequences include promoters, enhancers and other expression control elements (e.g., polyadenylation signals).
  • host cell is a cell that can be used to express a nucleic acid, e.g., a nucleic acid of the invention.
  • a host cell can be a prokaryote, for example, E. coli , or it can be an eukaryote, for example, a single-celled eukaryote (e.g., a yeast or other fungus), a plant cell (e.g., a tobacco or tomato plant cell), an animal cell (e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell) or a hybridoma.
  • a host cell is a cultured cell that can be transformed or transfected with a polypeptide-encoding nucleic acid, which can then be expressed in the host cell.
  • the phrase “recombinant host cell” can be used to denote a host cell that has been transformed or transfected with a nucleic acid to be expressed.
  • a host cell also can be a cell that comprises the nucleic acid but does not express it at a desired level unless a regulatory sequence is introduced into the host cell such that it becomes operably linked with the nucleic acid. It is understood that the term host cell refers not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to, e.g., mutation or environmental influence, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • Endothelin A receptor belongs to family A of 7-transmembrane receptors that are coupled to one or more intracellular signaling pathways via heterotrimeric guanine nucleotide-binding proteins (G proteins) (Jelinek et al., 1993 , Science 259:1614-1616, Segre et al., 1993 , Trends Endocrinol. Metab. 4:309-314).
  • G proteins heterotrimeric guanine nucleotide-binding proteins
  • the antibody provided herein can be selected to bind to membrane bound endothelin receptors as expressed on cells, and inhibit or block endothelin signaling through the endothelin receptors.
  • the antibody provided herein specifically binds to the human endothelin receptor.
  • the antibody binding to the human endothelin receptor can also bind to the endothelin receptors of other species, e.g., rat.
  • the examples below provide one method of generating murine antibodies which bind to human membrane-bound endothelin receptors, and in a further embodiment, bind to endothelin receptors of other species.
  • SEQ ID NO: 1-SEQ ID NO: 6 present sequences for human, monkey, and rat.
  • the sequence data were obtained from the GeneBank database of the National Center for Biotechnology Information.
  • Cynomolgus Homo sapiens ) polynucleotides (SEQ ID NO: 3); accession number: JV635771.
  • Cynomolgus Homo sapiens ) amino acid (SEQ ID NO: 4); accession number: AFJ71111.
  • Rat Rat ( Rattus norvegicus ) polynucleotides (SEQ ID NO: 5); accession number: M60786.
  • Rat Rat ( Rattus norvegicus ) amino acid (SEQ ID NO: 6); accession number: AAA41114.
  • an ET A R antibody for example, a full-length antibody, antibody fragment, antibody derivative, antibody variant, and antibody mutein.
  • the ET A R antibody provided herein comprises 1, 2, 3, 4, 5, or 6 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • Table 1 lists light chain CDR amino acid sequences of the ET A R antibodies provided herein, as well as their polynucleotides coding sequences.
  • Table 2 lists heavy chain CDR amino acid sequences of the ET A R antibodies provided herein, as well as their polynucleotides coding sequences.
  • the antibody provided herein comprises a sequence that differs from a CDR sequence listed in Table 1 and Table 2 by 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s). In another embodiment, the antibody provided herein comprises a sequence that differs from a CDR sequence listed in Table 1 and Table 2 by 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s). In yet another embodiment, the antibody provided herein comprises a sequence that differs from a CDR sequence listed in Table 1 and Table 2 by 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s).
  • the antibody provided herein comprises a sequence that differs from a CDR sequence listed in Table 1 and Table 2 by 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s). In still another embodiment, the antibody provided herein comprises a sequence that differs from a CDR sequence listed in Table 1 and Table 2 by 1 amino acid addition, substitution, and/or deletion.
  • ET A R antibody provided herein comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-1 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-1 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • ET A R antibody provided herein comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-2 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-2 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • ET A R antibody provided herein comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-3 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R-3 antibody further comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • the ET A R antibody provided herein comprises:
  • the ET A R antibody provided herein comprises a light chain CDR3 amino acid sequence independently selected from the list below: SEQ ID NO: 50, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 60, SEQ ID NO: 62, SEQ ID NO: 64, SEQ ID NO: 66, and SEQ ID NO: 68.
  • the ET A R antibody provided herein comprises a heavy chain CDR3 amino acid sequence independently selected from the list below: SEQ ID NO: 116, SEQ ID NO: 118, SEQ ID NO: 120, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 126, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 132, SEQ ID NO: 134, and SEQ ID NO: 136.
  • the ET A R antibody comprises a combination of light and heavy chain CDR3 amino acid sequences independently selected from the list below: SEQ ID NO: 50 versus SEQ ID NO: 116, SEQ ID NO: 62 versus SEQ ID NO: 128, SEQ ID NO: 62 versus SEQ ID NO: 130, SEQ ID NO: 64 versus SEQ ID NO: 132, SEQ ID NO: 66 versus SEQ ID NO: 134, and SEQ ID NO: 68 versus SEQ ID NO: 136.
  • the ET A R antibody comprises 1 or 2 amino acid sequences, wherein each amino acid sequence is independently selected from the amino acid sequences listed below:
  • a polynucleotide coding sequence of the ET A R antibody provided herein comprises 1 or 2 polynucleotide sequences, wherein each polynucleotide sequence is independently selected from the polynucleotide sequences listed below:
  • the ET A R antibody provided herein comprises:
  • the ET A R antibody provided herein comprises:
  • the ET A R antibody provided herein comprises a combination of amino acid sequences of light and heavy chain variable domains independently selected from the list below: SEQ ID NO: 138 and SEQ ID NO: 166 (L1H1), SEQ ID NO: 140 and SEQ ID NO: 168 (L2H2), SEQ ID NO: 142 and SEQ ID NO: 170 (L3H3), SEQ ID NO: 144 and SEQ ID NO: 172 (L4H4), SEQ ID NO: 146 and SEQ ID NO: 174 (L5H5), SEQ ID NO: 148 and SEQ ID NO: 176 (L6H6), SEQ ID NO: 150 and SEQ ID NO: 178 (L7H7), SEQ ID NO: 152 and SEQ ID NO: 180 (L8H8), SEQ ID NO: 154 and SEQ ID NO: 182 (L9H9), SEQ ID NO: 156 and SEQ ID NO: 184 (L10H10), SEQ ID NO: 158 and SEQ ID NO: 166
  • L2H1 refers to an antibody with a light chain variable region comprising the amino acid sequence of SEQ ID NO: 140 (L2) and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 166 (H1).
  • the ET A R antibody provided herein comprises a light chain variable domain selected from L1-L14 or a heavy chain variable domain selected from H1-H14, and fragments, derivatives, muteins, or variants thereof.
  • the ET A R antibody provided herein comprises a combination of light and heavy chain CDR3 amino acid sequences independently selected from the list below: SEQ ID NO: 138 and SEQ ID NO: 166, SEQ ID NO: 150 and SEQ ID NO: 178, SEQ ID NO: 152 and SEQ ID NO: 180, SEQ ID NO: 154 and SEQ ID NO: 182, SEQ ID NO: 156 and SEQ ID NO: 184, SEQ ID NO: 158 and SEQ ID NO: 186, SEQ ID NO: 160 and SEQ ID NO: 188, SEQ ID NO: 162 and SEQ ID NO: 190, and SEQ ID NO: 164 and SEQ ID NO: 192.
  • the ET A R antibody provided herein comprises light chain variable domain amino acid sequence SEQ ID NO: 138 or heavy chain variable domain amino acid sequence SEQ ID NO: 166. In another embodiment, the ET A R antibody provided herein comprises the combination of light chain variable domain amino acid sequence SEQ ID NO: 138 and heavy chain variable domain amino acid sequence SEQ ID NO: 166.
  • the ET A R antibody provided herein further comprises an amino acid sequence of a constant domain, wherein the amino acid sequence of the constant domain is independently selected from the amino acid sequences listed below:
  • the ET A R antibody provided herein further comprises constant domain amino acid sequences, wherein each constant domain amino acid sequence is independently selected from the combinations of the light chain and heavy chain constant domain amino acid sequences listed below:
  • the antibody provided herein comprises the amino acid sequences of the light and heavy chain CDRs and FRs (framework) illustrated above.
  • the antibody comprises a light chain CDR1 sequence illustrated above.
  • the antibody comprises a light chain CDR2 sequence illustrated above.
  • the antibody comprises a light chain CDR3 sequence illustrated above.
  • the antibody comprises a heavy chain CDR1 sequence illustrated above.
  • the antibody comprises a heavy chain CDR2 sequence illustrated above.
  • the antibody comprises a heavy chain CDR3 sequence illustrated above.
  • the antibody comprises a light chain FR1 sequence illustrated above.
  • the antibody comprises a light chain FR2 sequence illustrated above.
  • the antibody comprises a light chain FR3 sequence illustrated above. In another embodiment, the antibody comprises a light chain FR4 sequence illustrated above. In another embodiment, the antibody comprises a heavy chain FR1 sequence illustrated above. In another embodiment, the antibody comprises a heavy chain FR2 sequence illustrated above. In another embodiment, the antibody comprises a heavy chain FR3 sequence illustrated above. In another embodiment, the antibody comprises a heavy chain FR4 sequence illustrated above.
  • a CDR3 sequence of the antibody differs from the combination of SEQ ID NO: 50 and SEQ ID NO: 116 of the light chain and heavy chain CDR3 sequences illustrated above by no more than 6, 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s).
  • a light chain CDR3 sequence of the antibody differs from SEQ ID NO: 50 of the light chain CDR3 sequence illustrated above by no more than 6, 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s).
  • a light chain CDR3 sequence of the antibody differs from SEQ ID NO: 50 of the light chain CDR3 sequence illustrated above by no more than 6, 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s) and a heavy chain CDR3 sequence of the antibody differs from SEQ ID NO: 116 or SEQ ID NO: 118 of the heavy chain CDR3 sequence illustrated above by no more than 6, 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s).
  • the antibody further comprises a combination of 1, 2, 3, 4, 5 or 6 of light and heavy chain CDR sequences illustrated above.
  • the antibody further comprises a combination of 1, 2, 3, 4, 5 or 6 of light and heavy chain CDR sequences illustrated above, wherein each sequence independently differs from a combination of SEQ ID NO: 50 and SEQ ID NO: 116 of light chain and heavy chain CDR3 sequences by 6, 5, 4, 3, 2 or 1 amino acid addition(s), substitution(s), and/or deletion(s).
  • the antibody comprises the CDRs of a light chain variable region and the CDRs of a heavy chain variable region illustrated above.
  • the antibody comprises a combination of 1, 2, 3, 4, 5, and/or 6 of light and heavy chain CDR sequences illustrated above.
  • the antibody (such as an antibody or antibody fragment) comprises the amino acid sequence of light chain variable domain L1 illustrated above.
  • the sequence of the light chain variable domain differs from the sequence of light chain variable domain L1 by 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid deletion(s), insertion(s), or substitution(s).
  • the light-chain variable domain comprises an amino acid sequence at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97% or at least 99% identical to the sequence of light chain variable domain L1.
  • the polynucleotide coding sequence of the light chain variable domain comprises a nucleotide coding sequence at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97% or at least 99% identical to the polynucleotide coding sequence of L1.
  • the polynucleotide coding sequence of the light chain variable domain comprises a polynucleotide that hybridizes under moderately stringent conditions to the complement of the polynucleotide coding sequence of light chain variable domain L1.
  • the polynucleotide coding sequence of the light chain variable domain comprises a polynucleotide that hybridizes under stringent conditions to the complement of the polynucleotide coding sequence of light chain variable domain L1.
  • the antibody (such as an antibody or antibody fragment) comprises the amino acid sequence of heavy chain variable domain H1 illustrated above.
  • the sequence of the heavy chain variable domain differs from the sequence of heavy chain variable domain H1 by 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid deletion(s), insertion(s), or substitution(s).
  • the heavy chain variable domain comprises an amino acid sequence at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97% or at least 99% identical to the sequence of heavy chain variable domain H1.
  • a polynucleotide coding sequence of the heavy chain variable domain comprises a polynucleotide sequence at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97% or at least 99% identical to the polynucleotide sequence of H1.
  • the polynucleotide coding sequence of the heavy chain variable domain comprises a polynucleotide that hybridizes under moderately stringent conditions to the complement of a polynucleotide coding sequence of the heavy chain variable domain H1.
  • the polynucleotide coding sequence of the heavy chain variable domain comprises a polynucleotide that hybridizes under stringent conditions to the complement of a polynucleotide coding sequence of the heavy chain variable domain H1.
  • the antibody provided herein include the combination of L1H1, L2H2, L3H3, L4H4, L5H5, L6H6, L7H7, L8H8, L9H9, L10H10, L11H11, L12H12, L13H13 or L14H14; or an isotype thereof (for example, IgA, IgG1, IgG2a, IgG2b, IgG3, IgM, IgE, or IgD) or a Fab or F(ab′) 2 fragment thereof.
  • the antibody provided herein includes an antibody comprising a combination of L1H1, or a converted isotype antibody thereof (for example, IgA, IgG1, IgG2a, IgG2b, IgG3, IgM, IgE, or IgD) or a Fab or F(ab′) 2 fragment thereof.
  • a converted isotype antibody thereof for example, IgA, IgG1, IgG2a, IgG2b, IgG3, IgM, IgE, or IgD
  • a Fab or F(ab′) 2 fragment thereof for example, IgA, IgG1, IgG2a, IgG2b, IgG3, IgM, IgE, or IgD
  • the antibody e.g., an antibody, antibody fragment, and antibody derivative
  • the light chain constant region can be, for example, a kappa- or lambda-type light chain constant region, e.g., a murine kappa- or lambda-type light chain constant region.
  • the heavy chain constant region can be, for example, an alpha-, delta-, epsilon-gamma-, or mu-type heavy chain constant regions, e.g., a murine alpha-, delta-, epsilon-, gamma-, or mu-type heavy chain constant region.
  • the light or heavy chain constant region is a fragment, derivative, variant, or mutein of a naturally occurring constant region.
  • the antibody provided herein further comprises a constant light chain ⁇ or ⁇ region or a fragment thereof.
  • the light chain constant region sequence and its polynucleotide coding sequence are provided as follows:
  • the antibody provided herein further comprises a heavy chain constant region or a fragment thereof.
  • the heavy chain constant region sequence and its polynucleotide coding sequence are provided as follows:
  • polynucleotide (IgG1), (SEQ ID NO: 197); amino acid (IgG1), (SEQ ID NO: 198)
  • the ET A R antibody provided herein is selected from murine antibodies, humanized antibodies, chimeric antibodies, monoclonal antibodies, polyclonal antibodies, recombinant antibodies, antigen-binding antibody fragments, single-chain antibodies, double-chain antibodies, triple-chain antibodies, tetra-chain antibodies, Fab fragments, F(ab′)x fragments, domain antibodies, IgD antibodies, IgE antibodies, IgM antibodies, IgG1 antibodies, IgG2 antibodies, IgG3 antibodies, and IgG4 antibodies.
  • the ET A R antibody provided herein is an ET A R monoclonal antibody.
  • the ET A R antibody provided herein is a mouse ET A R antibody.
  • the ET A R antibody provided herein is a humanized ET A R antibody.
  • the ET A R antibody provided herein is monoclonal antibody A-1 (comprising SEQ ID NO: 138 and SEQ ID NO: 166), A-7 (comprising SEQ ID NO: 150 and SEQ ID NO: 178), A-8 (comprising SEQ ID NO: 152 and SEQ ID NO: 180), A-9 (comprising SEQ ID NO: 154 and SEQ ID NO: 182), A-10 (comprising SEQ ID NO: 156 and SEQ ID NO: 184), A-11 (comprising SEQ ID NO: 158 and SEQ ID NO: 186), A-12 (comprising SEQ ID NO: 160 and SEQ ID NO: 188), A-13 (comprising SEQ ID NO: 162 and SEQ ID NO: 190), or A-14 (comprising SEQ ID NO: 164 and SEQ ID NO: 192).
  • A-1 comprising SEQ ID NO: 138 and SEQ ID NO: 166
  • A-7 comprising SEQ ID NO: 150 and SEQ
  • the antibody provided herein is a full-length antibody (including polyclonal, monoclonal, chimeric, humanized or human antibody with full length heavy and/or light chains).
  • the antibody provided herein is an antibody fragment, for example, F(ab′) 2 , Fab, Fab′, Fv, Fc, or Fd fragment, and can be incorporated into single domain antibodies, single-chain antibodies, maxibodies, minibodies, intrabodies, double-chain antibodies, triple-chain antibodies, tetra-chain antibodies, v-NAR and bis-scFv (see e.g., Hollinger and Hudson, 2005 , Nature Biotechnology, 23, 9, 1126-1136).
  • the antibody provided herein also includes antibody polypeptides such as those disclosed in U.S. Pat. No. 6,703,199, including fibronectin polypeptide monobodies.
  • the antibody provided herein also includes other antibody polypeptides disclosed in U.S. Patent Publication 2005/0238646, which are single-chain polypeptides.
  • variable regions of a gene expressing a monoclonal antibody of interest are amplified using nucleotide primers in a hybridoma.
  • nucleotide primers can be synthesized by one of ordinary skill in the art, or can be purchased from commercially available sources (see, e.g., Stratagene, La Jolla, Calif.), which sells primers for mouse and human variable regions including, among others, primers for V Ha , V Hb , V Hc , V Hd , C H1 , V L and C L regions.
  • These primers can be used to amplify heavy or light chain variable regions, which can then be inserted into vectors such as IMMUNOZAPTMH or IMMUNOZAPTML (Stratagene), respectively.
  • vectors can then be introduced into E. coli , yeast, or mammalian-based systems for expression. Large amounts of a single-chain protein containing a fusion of the V H and V L regions can be produced using these methods (see Bird el al., 1988 , Science 242:423-426).
  • the genes of the specific antibodies can be cloned by isolating and amplifying DNA or mRNA therefrom according to standard procedures described herein.
  • the antibodies produced therefrom can be sequenced to identify CDRs, and the coding DNA of the CDRs can be manipulated as described above to generate other antibodies provided herein.
  • Antibodies provided herein preferably modulate endothelin signaling in the cell-based assay described herein and/or in the in vivo assay described herein and/or cross-block the binding of one of the antibodies described herein and/or are cross-blocked from binding ET A R by one of the antibodies described herein. Accordingly, such binding agents can be identified using the assays described herein.
  • antibodies are generated by first identifying antibodies that bind to cells overexpressing ET A R and/or neutralize in the cell-based and/or in vivo assays described herein and/or cross-block the antibodies described herein and/or are cross-blocked from binding ET A R by one of the antibodies described herein.
  • An alternative method for production of a murine monoclonal antibody is to inject hybridoma cells into the peritoneal cavity of a syngeneic mouse, for example, a mouse that has been treated (e.g., pristine-primed) to promote formation of ascites fluid containing the monoclonal antibody.
  • Monoclonal antibodies can be isolated and purified by a variety of well-established techniques.
  • Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, e.g., Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al., “Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)).
  • a monoclonal antibody can be purified by affinity chromatography using an appropriate ligand selected based on particular properties of the antibody (e.g., heavy or light chain isotype, binding specificity, etc.).
  • suitable ligands immobilized on a solid support include Protein A, Protein an anti-constant region (light chain or heavy chain) antibody, an anti-idiotype antibody, and a TGF- ⁇ binding protein, or a fragment or variant thereof.
  • CDRs complementarity determining regions
  • Antibodies against human endothelin A receptor can be used, for example, in assays to detect the presence of the endothelin A receptor, either in vitro or in vivo.
  • Antibodies can also be prepared by any of the conventional techniques. For example, they can be purified from cells that naturally express them (e.g., an antibody can be purified from a hybridoma that produces it) or produced in recombinant expression systems using any technique known in the art. See, for example, Monoclonal Antibodies, Hybridomas: A New Dimension in Biological Analyses, Kennet et al. (eds.), Plenum Press, New York (1980); and Antibodies: A Laboratory Manual, Harlow and Land (eds.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1988). This is discussed in the nucleic acid section below.
  • Antibodies can be prepared and screened for desired properties by any known techniques. Some techniques relate to the isolation of nucleic acids encoding polypeptide chains (or portions thereof) of related antibodies (e.g., anti-ET A R antibodies) and manipulation of nucleic acid. Nucleic acids can be fused with another relevant nucleic acid or modified by recombinant DNA techniques (e.g., induced mutations or other conventional techniques) to add, delete or replace one or more amino acid residues.
  • recombinant DNA techniques e.g., induced mutations or other conventional techniques
  • such antibodies can be obtained by a number of affinity maturation protocols, including maintaining the CDRs (Yang et al., 1995 , J. Mol. Biol., 254:392-403), chain shuffling (Marks et al., 1992 , Bio/Technology, 10:779-783), use of mutation strains of E. coli . (Low et al., 1996 , J. Mol. Biol., 250:350-368), DNA shuffling (Patten et al., 1997 , Curr. Opin.
  • fragments of the ET A R antibody are provided herein. Such fragments can comprise entirely antibody-derived sequences or additional sequences. Examples of antigen binding fragments include Fab, F(ab′) 2 , single chain antibodies, diabodies, tribodies, tetrabodies, and domain antibodies. Other examples are provided in Lunde et al., 2002 , Biochem. Soc. Trans. 30:500-06.
  • Single chain antibodies can be formed by linking heavy and light chain variable domain (Fv region) fragments via an amino acid bridge (short peptide linker), resulting in a single polypeptide chain.
  • Fv region heavy and light chain variable domain
  • short peptide linker short peptide linker
  • Such single-chain Fvs have been prepared by fusion DNA encoding a peptide linker between DNAs encoding the two variable domain polypeptides (V L and V H ).
  • the resulting polypeptides can fold back on themselves to form antigen-binding monomers, or they can form multimers (e.g., dimers, trimers, or tetramers), depending on the length of a flexible linker between the two variable domains (Kortt et al., 1997 , Prot. Eng.
  • Single chain antibodies derived from antibodies provided herein including, but not limited to, scFvs comprising the variable domain combination L1H1, are encompassed by the present invention.
  • Antibodies derived from an antibody can also be obtained, for example, by proteolytic hydrolysis of the antibody, for example, pepsin or papain digestion of a whole antibody according to conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a SS fragment termed F(ab′) 2 . This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab′ monovalent fragments.
  • the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages.
  • CDRs complementarity determining regions
  • Another form of an antibody fragment is a peptide comprising one or more complementarity determining regions (CDRs) of an antibody.
  • CDRs can be obtained by constructing polynucleotides that encode the CDRs.
  • Such polynucleotides are prepared, for example, by using the polymerase chain reaction to synthesize the variable region using mRNA or antibody-producing cells as a template (see, for example, Larrick et al., 1991, Methods: A Companion to Methods in Enzymology 2:106; Courtenay-Luck, “(Genetic Manipulation of Monoclonal Antibodies,” in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al.
  • the antibody fragment further can comprise at least one variable region domain of an antibody described herein.
  • the V region domain can be monomeric and be a V H or V L domain, which can bind to ET A R with an affinity of 1 ⁇ 10 ⁇ 7 M or less as described below.
  • variable region domain can be any naturally occurring variable domain or an engineered version thereof.
  • engineered version is meant a variable region domain that has been created using recombinant DNA engineering techniques.
  • engineered versions include those created, for example, from a specific antibody variable region by insertions, deletions, or changes in or to the amino acid sequences of the specific antibody.
  • Particular examples include engineered variable region domains containing at least one CDR and optionally one or more framework amino acids from a first antibody and the remainder of the variable region domain from a second antibody.
  • variable region domain can be covalently attached at a C-terminal amino acid to at least one other antibody domain or a fragment thereof.
  • a V H domain that is present in the variable region domain can be linked to an immunoglobulin C H1 domain or a fragment thereof.
  • a V L domain can be linked to a C K domain or a fragment thereof.
  • the antibody can be a Fab fragment, wherein the antigen binding domain contains associated V H and V L domains covalently linked at their C-termini to a C H1 and C ⁇ domain, respectively.
  • the C H1 domain can be extended with further amino acids, for example to provide a hinge region or a portion of a hinge region domain as found in a Fab′ fragment, or to provide further domains, such as antibody C H2 and C H3 domains.
  • nucleotide sequences of L1 and H1, encoding the corresponding amino acid sequences of A-1 can be altered, for example, by random mutagenesis or by site-directed mutagenesis (e.g., oligonucleotide-directed site-specific mutagenesis) to create an altered polynucleotide comprising one or more particular nucleotide substitutions, deletions, or insertions as compared to the non-mutated polynucleotide.
  • site-directed mutagenesis e.g., oligonucleotide-directed site-specific mutagenesis
  • anti-endothelin receptor antibodies within the scope or this invention include covalent or aggregative conjugates or anti-endothelin receptor antibodies, or fragments thereof, with other proteins or polypeptides, such as by expression or recombinant fusion proteins comprising heterologous polypeptides fused to the N-terminus or C-terminus or an anti-endothelin receptor antibody polypeptide.
  • the conjugated peptide can be a heterologous signal (or leader) polypeptide, e.g., the yeast alpha-factor leader or a peptide such as an epitope tag.
  • An antibody containing fusion proteins can comprise peptides added to facilitate purification or identification of antigen binding protein (e.g., poly-His).
  • an antibody also can be linked to the FLAG peptide as described in Hopp et al., 1988 , Bio/Technology 6:1204, and U.S. Pat. No. 5,011,912.
  • the FLAG peptide is highly antigenic and provides an epitope reversibly bound by a specific monoclonal antibody (mAb), enabling rapid assay and facile purification of an expressed recombinant protein.
  • mAb monoclonal antibody
  • Reagents useful for preparing fusion proteins in which the FLAG peptide is fused to a given polypeptide are commercially available (Sigma, St. Louis, Mo.).
  • oligomers that contain one or more antibodies can be employed as endothelin receptor antagonists.
  • Oligomers can be in the form of covalently-linked or non-covalently-linked dimers, trimers, or higher oligomers. Oligomers comprising two or more antibodies are contemplated for use, with one example being a homodimer. Other oligomers include heterodimers, homotrimers, heterotrimers, homotetramers, heterotetramers, etc.
  • One embodiment is directed to oligomers comprising multiple antibodies joined via covalent or non-covalent interactions between peptide moieties fused to the antibodies.
  • Such peptides can be peptide linkers (spacers), or peptides that have the property of promoting oligomerization.
  • Leucine zippers and certain polypeptides derived from antibodies are among the peptides that can promote oligomerization of antibodies attached thereto, as described in more detail below.
  • the oligomers comprise from two to four antibodies.
  • the antibodies of the oligomer can be in any form, such as any of the forms described above, e.g., variants or fragments.
  • the oligomers comprise antibodies that have endothelin receptor binding activity.
  • an oligomer is prepared using polypeptides derived from immunoglobulins.
  • Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., 1991 , PNAS USA 88:10535; Byrn et al., 1990 , Nature 344:677; and Hollenbaugh et al., 1992 “Construction of Immunoglobulin Fusion Proteins”, in Current Protocols in Immunology, Suppl. 4, pages 10.19.1-10.19.11.
  • One embodiment provided herein is directed to a dimer comprising two fusion proteins created by fusing an endothelin receptor binding fragment of an anti-endothelin A receptor antibody to the Fc region of an antibody.
  • the dimer can be made by, for example, inserting a gene fusion encoding the fusion protein into an appropriate expression vector, expressing the gene fusion in host cells transformed with the recombinant expression vector, and allowing the expressed fusion protein to assemble much like antibody molecules, whereupon inter-chain disulfide bonds form between the Fc moieties to yield the dimer.
  • Fc polypeptide as used herein includes native and mutein forms of polypeptides derived from the Fc region of an antibody. Truncated forms of such polypeptides containing the hinge region that promotes dimerization also are included. Fusion proteins comprising Fc moieties (and oligomers formed therefrom) offer the advantage of facile purification by affinity chromatography over Protein A or Protein G columns.
  • Fc polypeptide is a single chain polypeptide extending from the N-terminal hinge region to the native C-terminus of the Fc region of a human IgG1 antibody.
  • Another useful Fc polypeptide is the Fc mutein described in U.S. Pat. No. 5,457,035 and in Baum et al., 1994 , EMBO J. 13:3992-4001.
  • the amino acid sequence of this mutein is identical to that of the native Fc sequence presented in WO 93/10151, except that amino acid 19 has been changed from Leu to Ala, amino acid 20 has been changed from Leu to Glu, and amino acid 22 has been changed from Gly to Ala.
  • the mutein exhibits reduced affinity for Fc receptors.
  • the variable portion of the heavy and/or light chains of an anti-endothelin receptor antibody can be substituted for the variable portion of an antibody heavy and/or light chain.
  • the oligomer is a fusion protein comprising multiple antibodies, with or without peptide linkers (spacer peptides).
  • suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233.
  • Leucine zipper domains are peptides that promote oligomerization of the proteins in which they are found.
  • Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., 1988 , Science 240:1759), and have since been found in a variety of different proteins.
  • the known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize.
  • leucine zipper domains suitable for producing soluble oligomeric proteins are described in PCT application WO 94/10308, and the leucine zipper derived from lung surfactant protein D (SPD) described in Hoppe et al., 1994 , FEBS Letters 344:191, hereby incorporated by reference.
  • SPD lung surfactant protein D
  • the use of a modified leucine zipper that allows for stable trimerization of a heterologous protein fused thereto is described in Fanslow et al., 1994 , Semin. Immunol. 6:267-78.
  • recombinant fusion proteins comprising an anti-endothelin receptor antibody fragment or derivative fused to a leucine zipper peptide are expressed in suitable host cells, and the soluble oligomeric anti-endothelin receptor antibody fragments or derivatives that form are recovered from the culture supernatant.
  • the antibody derivatives can comprise at least one of the CDRs disclosed herein.
  • one or more CDR can be incorporated into known antibody framework regions (IgG1, IgG2, etc.), or conjugated to a suitable vehicle to enhance the half-life thereof.
  • suitable vehicles include, but are not limited to Fc, albumin, transferrin, and the like. These and other suitable vehicles are known in the art.
  • conjugated CDR peptides can be in monomeric, dimeric, tetrameric, or other form.
  • one or more water-soluble polymer is bonded at one or more specific position, for example at the amino terminus, of a binding agent.
  • an antibody derivative comprises one or more water soluble polymer attachments, including, but not limited to, polyethylene glycol, polyoxyethylene glycol, or polypropylene glycol. See, e.g., U.S. Pat. Nos. 4,640,835, 4,496,689, 4,301,144, 4,670,417, 4,791,192 and 4,179,337.
  • a derivative comprises one or more of monomethoxy-polyethylene glycol, dextran, cellulose, or other carbohydrate based polymers, poly-(N-vinyl pyrrolidone)-polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well as mixtures of such polymers.
  • one or more water-soluble polymer is randomly attached to one or more side chains.
  • PEG can act to improve the therapeutic capacity for a binding agent, such as an antibody. Certain such methods are discussed, for example, in U.S. Pat. No. 6,133,426, which is hereby incorporated by reference for any purpose.
  • an antibody provided herein can have at least one amino acid substitution, providing that the antibody retains binding specificity. Therefore, modifications to the antibody structures are encompassed within the scope of the invention. These can include amino acid substitutions, which may be conservative or non-conservative, that do not destroy the human endothelin receptor binding capability of an antibody. Conservative amino acid substitutions may encompass non-naturally occurring amino acid residues, which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. These include peptidomimetics and other reversed or inverted forms of amino acid moieties.
  • a conservative amino acid substitution can also involve a substitution of a native amino acid residue with a normative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position.
  • Non-conservative substitutions can involve the exchange of a member of one class of amino acids or amino acid mimetics for a member from another class with different physical properties (e.g., size, polarity, hydrophobicity, charge).
  • variants to be tested may be generated, which contain a single amino acid substitution at each desired amino acid residue.
  • the variants can then be screened using activity assays known to those skilled in the art.
  • Such variants could be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed, undesirably reduced, or unsuitable activity, variants with such a change may be avoided.
  • one skilled in the art can readily determine the amino acids where further substitutions should be avoided either alone or in combination with other mutations.
  • a skilled artisan will be able to determine suitable variants of the polypeptide as set forth herein using well-known techniques.
  • one skilled in the art may identify suitable areas of the molecule that may be changed without destroying activity by targeting regions not to be important for activity.
  • even areas that may be important for biological activity or for structure may be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure.
  • one skilled in the art can review structure-function studies identifying residues in similar polypeptides that are important for activity or structure.
  • One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar polypeptides. In view of such information, one skilled in the art may predict the alignment of amino acid residues of an antibody with respect to its three dimensional structure. In certain embodiments, one skilled in the art may choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules.
  • a number of scientific publications have been devoted to the prediction of secondary structure. See Moult, 1996 , Curr. Op. Biotech.
  • Additional methods of predicting secondary structure include “threading” (Jones, 1997 , Curr. Opin. Struct. Biol. 7:377-87; Sippl et al., 1996 , Structure 4:15-19), “profile analysis” (Bowie et al., 1991 , Science 253:164-170; Gribskov et al., 1990 , Meth. Enzym. 183:146-159; Gribskov et al., 1987 , Proc. Nat. Acad. Sci. 84:4355-4358), and “evolutionary linkage” (see Holm, supra (1999), and Brenner, supra (1997)).
  • variants of antibodies include glycosylation variants, wherein the number and/or type of glycosylation sites have been altered compared to the amino acid sequences of a parent polypeptide.
  • variants comprise a greater or lesser number of N-linked glycosylation sites than the native protein.
  • elimination of such a sequence by substitutions removes an existing N-linked carbohydrate chain.
  • rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created.
  • Additional preferred antibody variants include cysteine variants, wherein one or more cysteine residues are deleted from or substituted for another amino acid (e.g., serine) as compared to the parent amino acid sequence.
  • cysteine variants can be useful when antibodies must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.
  • amino acid substitutions can be used to identify important residues of antibodies to human endothelin receptor, or to increase or decrease the affinity of the antibodies to human endothelin receptor described herein.
  • preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and/or (4) confer or modify other physiochemical or functional properties on such polypeptides.
  • single or multiple amino acid substitutions in certain embodiments, conservative amino acid substitutions can be made in the naturally-occurring sequence (in certain embodiments, in the portion of the polypeptide outside the domain(s) forming intermolecular contacts).
  • a conservative amino acid substitution typically cannot substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • a replacement amino acid should not break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence.
  • Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (Branden and Tooze, Eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et al., 1991 , Nature 354:105, each of which is incorporated herein by reference.
  • antibodies of the invention can be chemically bonded with polymers, lipids, or other moieties.
  • the antigen binding agents can comprise at least one of the CDRs described herein incorporated into a biocompatible framework structure.
  • the biocompatible framework structure comprises a polypeptide or portion thereof that is sufficient to form a conformationally stable structural support, or framework, or scaffold, which is able to present one or more sequences of amino acids that bind to an antigen (e.g., CDRs, a variable region, etc.) in a localized surface region.
  • an antigen e.g., CDRs, a variable region, etc.
  • Such structures can be a naturally occurring polypeptide or polypeptide “fold” (a structural motif), or can have one or more modifications, such as additions, deletions or substitutions of amino acids, relative to a naturally occurring polypeptide or fold.
  • These scaffolds can be derived from a polypeptide of any species (or of more than one species), such as a human, other mammal, other vertebrate, invertebrate, plant, bacteria or virus.
  • the biocompatible framework structures are based on protein scaffolds or skeletons other than immunoglobulin domains.
  • protein scaffolds or skeletons other than immunoglobulin domains.
  • those based on fibronectin, ankyrin, lipocalin, neocarzinostain, cytochrome b, CP1 zinc finger, PST1, coiled coil, LACI-D1, Z domain and tendamistat domains can be used (see, e.g., Nygren and Uhlen, 1997 , Current Opinion in Structural Biology 7:463-469).
  • suitable binding agents include portions of these antibodies, such as one or more of heavy chain CDR1, CDR2, CDR3, light chain CDR1, CDR2 and CDR3 as specifically disclosed herein. At least one of the regions of heavy chain CDR1, CDR2, CDR3, CDR1, CDR2 and CDR3 can have at least one amino acid substitution, provided that the antibody retains the binding specificity of the non-substituted CDR.
  • the non-CDR portion of the antibody can be a non-protein molecule, wherein the binding agent cross-blocks the binding of an antibody disclosed herein to human endothelin receptor and/or inhibits the activity of endothelin-1 signaling through the receptor.
  • the non-CDR portion of the antibody can be a non-protein molecule in which the antibody exhibits a similar binding pattern to human endothelin receptor peptides in a competition binding assay as that exhibited by at least one of antibodies A1/A2, and/or neutralizes the activity of endothelin-1.
  • the non-CDR portion of the antibody can be composed of amino acids, wherein the antibody is a recombinant binding protein or a synthetic peptide, and the recombinant binding protein cross-blocks the binding of an antibody disclosed herein to human ET A R and/or neutralizes endothelin-1 activity in vitro or in vivo.
  • the non-CDR portion of the antibody can be composed of amino acids, wherein the antibody is a recombinant antibody, and the recombinant antibody exhibits a similar binding pattern to human ET A R peptides in a competition binding assay as exhibited by at least one of the antibodies A1/A2, and/or neutralizes endothelin-1 signaling.
  • the present invention provides isolated nucleic acid molecules that encode the antibodies provided herein.
  • the nucleic acids comprise, for example, polynucleotides that encode all or part of an antibody, for example, one or both chains of an antibody of the invention, or a fragment, derivative, mutein, or variant thereof; polynucleotides sufficient for use as hybridization probes; PCR primers or sequencing primers for identifying, analyzing, mutating or amplifying a polynucleotide encoding a polypeptide; anti-sense nucleic acids for inhibiting expression of a polynucleotide, and complementary sequences of the foregoing.
  • he nucleic acids can be any length.
  • nucleic acids can be single-stranded or double-stranded and can comprise RNA and/or DNA nucleotides, and artificial variants thereof (e.g., peptide nucleic acids).
  • Nucleic acids encoding antibody polypeptides can be isolated from B-cells of mice that have been immunized with ET A R antigen.
  • the nucleic acid can be isolated by conventional procedures such as polymerase chain reaction (PCR).
  • nucleic acid sequences encoding the variable regions of the heavy and light chain variable regions are shown above. The skilled artisan will appreciate that, due to the degeneracy of the genetic code, each of the polypeptide sequences disclosed herein is encoded by a large number of other nucleic acid sequences. The present invention provides each degenerate nucleotide sequence encoding each antibody of the invention.
  • the invention further provides nucleic acids that hybridize to other nucleic acids (e.g., nucleic acids comprising a nucleotide sequence of any of A-1/A-2) under particular hybridization conditions.
  • nucleic acids e.g., nucleic acids comprising a nucleotide sequence of any of A-1/A-2
  • Methods for hybridizing nucleic acids are well-known in the art. See, e.g., Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • a moderately stringent hybridization condition uses a prewashing solution containing 5 ⁇ sodium chloride/sodium citrate (SSC), 0.5% SDS, 1.0 mM EDTA (pH 8.0), hybridization buffer of about 50% formamide, 6 ⁇ SSC, and a hybridization temperature of 55° C.
  • hybridization and/or washing conditions can manipulate the hybridization and/or washing conditions to increase or decrease the stringency of hybridization such that nucleic acids comprising nucleotide sequences that are at least 65, 70, 75, 80, 85, 90, 95, 98 or 99% identical to each other typically remain hybridized to each other.
  • Mutations can be introduced using any technique known in the art.
  • one or more particular amino acid residues are changed using, for example, a site-directed mutagenesis protocol.
  • one or more randomly selected residues is changed using, for example, a random mutagenesis protocol. No matter how it is made, a mutant polypeptide can be expressed and screened for a desired property.
  • nucleic acid without significantly altering the biological activity of a polypeptide that it encodes. For example, one can make nucleotide substitutions leading to amino acid substitutions at non-essential amino acid residues.
  • nucleotide sequences provided herein for L1 to L2 and E11 to H2, or fragments, variants, or derivatives thereof are mutated such that they encode amino acid sequences provided herein for L1 to L2 and E11 to H2, comprising one or more deletions or substitutions of amino acid residues to result in sequences bearing two or more different amino acid residues.
  • the mutagenesis inserts an amino acid adjacent to one or more amino acid residues shown herein for L1 to L2 and E11 to H2 to result in sequences with two or more different amino acid residues.
  • one or more mutations can be introduced into a nucleic acid that selectively change the biological activity. (e.g., binding to ET A R) of a polypeptide that it encodes.
  • the mutation can quantitatively or qualitatively change the biological activity. Examples of quantitative changes include increasing, reducing or eliminating the activity. Examples of qualitative changes include changing the antigen specificity of an antibody.
  • the present invention provides nucleic acid molecules that are suitable for use as primers or hybridization probes for the detection of nucleic acid sequences of the invention.
  • a nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence encoding a full-length polypeptide of the invention, for example, a fragment that can be used as a probe or primer or a fragment encoding an active portion (e.g., a ET A R binding portion) of a polypeptide of the invention.
  • Probes based on the sequence of a nucleic acid of the invention can be used to detect the nucleic acid or similar nucleic acids, for example, transcripts encoding a polypeptide of the invention.
  • the probe can comprise a label group, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used to identify a cell that expresses the polypeptide.
  • the vectors provided herein comprise a nucleic acid encoding a polypeptide of the invention or a portion thereof.
  • vectors include, but are not limited to, plasmids, viral vectors, non-episomal mammalian vectors and expression vectors, for example, recombinant expression vectors.
  • the recombinant expression vectors provided herein can comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell.
  • the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed.
  • Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cells (e.g., SV40 early gene enhancer, Rous sarcoma virus promoter and cytomegalovirus promoter), those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences, see Voss et al., 1986 , Trends Biochem. Sci.
  • the present invention provides host cells into which a recombinant expression vector of the invention has been introduced.
  • a host cell can be any prokaryotic cell or eukaryotic cell.
  • Prokaryotic host cells include gram negative or gram positive organisms, for example, E. coli or bacilli.
  • Higher eukaryotic cells include insect cells, yeast cells, and established cell lines of mammalian origin.
  • suitable mammalian host cell lines include Chinese hamster ovary (CHO) cells or their derivatives such as Veggie CHO and related cell lines which grow in serum-free media (see Rasmussen et al., 1998 , Cytotechnology 28:31) or CHO strain DXB-11, which is deficient in DHFR (see Urlaub et al., 1980 , Proc. Natl. Acad. Sci. USA 77:4216-20).
  • Additional CHO cell lines include CHO-K1 (ATCC#CCL-61), EM9 (ATCC# CRL-1861), and W20 (ATCC# CRL-1862).
  • Additional host cells include the COS-7 line of monkey kidney cells (ATCC# CRL-1651) (see Gluzman et al., 1981 , Cell 23:175), L cells, C127 cells, 3T3 cells (ATCC CCL-163), AM-1/D cells (described in U.S. Pat. No. 6,210,924), HeLa cells, BHK (ATCC CRL-10) cell lines, the CV1/EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL-70) (see McMahan et al., 1991 , EMBO J.
  • human embryonic kidney cells such as 293, 293 EBNA or MSR 293, human epidermal A431 cells, human Colo205 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HL-60, U937, HaK or Jurkat cells.
  • Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985).
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • a gene that encodes a selectable marker e.g., for resistance to antibiotics
  • Preferred selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die), among other methods.
  • the transformed cells can be cultured under conditions that promote expression of a polypeptide, and the polypeptide recovered by conventional protein purification procedures.
  • One such purification procedure is described in the Examples below.
  • Polypeptides contemplated for use herein include substantially homogeneous recombinant mammalian anti-endothelin receptor antibody polypeptides substantially free of contaminating endogenous materials.
  • the antibody provided herein specifically binds to an endothelin receptor, inhibits the signaling transduction, and demonstrates a therapeutic biological effect, for example, the attenuation of pulmonary arterial hypertension in an animal model.
  • a mouse or humanized antibody provided herein specifically binds to a human endothelin receptor.
  • Such an antibody includes an antagonistic or neutralizing antibody that reduces or neutralizes endothelin signaling.
  • the K d of the antibody provided herein binding to a human endothelin receptor ET A R is ranging approximately from 0.01 nM to 1000 nM, from 0.1 nM to 500 nM, from 0.5 nM to 200 nM, from 1 nM to 200 nM, or from 10 nM to 100 nM.
  • the K d of the antibody provided herein binding to a human endothelin receptor ET A R is approximately from 1 nM to 200 nM.
  • the K d of the antibody provided herein binding to a human endothelin receptor ET A R is approximately from 10 nM to 100 nM.
  • the K d of the antibody provided herein binding to a human endothelin receptor ET A R is approximately 1 nM, 2 nM, 5 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, or 100 nM.
  • the IC 50 of the antibody provided herein antagonizing endothelin signaling is ranging approximately from 0.01 nM to 500 nM, from 0.1 nM to 200 nM, from 0.5 nM to 200 nM, from 1 nM to 200 nM, or from 10 nM to 100 nM. In another embodiment, the IC 50 of the antibody provided herein antagonizing endothelin signaling is approximately from 1 nM to 200 nM. In yet another embodiment, the IC 50 of the antibody provided herein antagonizing endothelin signaling is approximately from 10 nM to 100 nM.
  • the IC 50 of the antibody provided herein antagonizing endothelin signaling is approximately 1 nM, 2 nM, 5 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, or 100 nM.
  • the antibody provided herein specifically binds to a human endothelin receptor ET A R with one or more following properties:
  • the reference antibody comprises a combination of light chain variable domain amino acid sequence SEQ ID NO: 138 and heavy chain variable domain amino acid sequence SEQ ID NO: 166.
  • the reference antibody is monoclonal antibody A-1, A-2, A-7, A-9, or A-12.
  • the term “substantially similar” means comparable to, or approximately 100%, 99%, 98%, 97%, 95%, 90%, 85%, 80%, 75%, 70%, 65%, or 50% identical to the IC 50 or Kb (or K d ) of a reference antibody.
  • the reference antibody is, for example, an antibody comprising a heavy chain and light chain combination L1H1 or L2H2.
  • the reference antibody includes A-1.
  • the ET A R antibody provided herein is able to bind to a human endothelin receptor specifically and lower pulmonary arterial hypertension in an animal model.
  • the pulmonary arterial hypertension is lowered by about 2% compared with an animal without treatment.
  • the pulmonary arterial hypertension is lowered by about 5% compared with an animal without treatment.
  • the pulmonary arterial pressure is lowered by about 10% compared to an animal without treatment.
  • the pulmonary arterial hypertension is lowered by about 15% compared to an animal without treatment.
  • the pulmonary arterial hypertension is lowered by about 20% compared to an animal without treatment.
  • the pulmonary arterial hypertension is lowered by about 25% compared to an animal without treatment.
  • the amount of reduction of pulmonary arterial hypertension is controlled by dosage.
  • a therapeutically effective dosage is the dosage required to reduce pulmonary arterial hypertension into the normal range for an animal or human patient.
  • the pharmaceutical composition provided herein comprises an ET A R antibody provided herein and one or more pharmaceutically acceptable carriers.
  • a stable pharmaceutical solution formulation of the ET A R antibody is provided herein.
  • the stable pharmaceutical solution formulation of the ET A R antibody is stable and efficacious with a longer half-life in vivo, and can be used to effectively treat pulmonary arterial hypertension and related diseases and cancer of a reproductive organ.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein comprises an ET A R antibody provided herein and a buffer.
  • the pH of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is ranging approximately from 4 to 11, from 5 to 7, or from 5 to 6.
  • the pH of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is approximately from 5 to 7.
  • the pH of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is approximately from 5 to 6.
  • the pH of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is approximately from 5.3 to 6.5.
  • the pH of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is about 5.8.
  • the concentration of the ET A R antibody provided herein is approximately ranging from 10 to 500 mg/mL, from 10 to 250 mg/mL, from 10 to 200 mg/mL, or from 10 to 100 mg/mL.
  • the concentration of the ET A R antibody provided herein is approximately 10 mg/mL, 15 mg/mL, 20 mg/mL, 25 mg/mL, 30 mg/mL, 35 mg/mL, 40 mg/mL, 45 mg/mL, 50 mg/mL, 55 mg/mL, 60 mg/mL, 65 mg/mL, 70 mg/mL, 75 mg/mL, 80 mg/mL, 85 mg/mL, 90 mg/mL, 95 mg/mL, or 100 mg/mL.
  • the concentration of the ET A R antibody provided herein is approximately 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, 150 mg/mL, 160 mg/mL, 170 mg/mL, 180 mg/mL, 190 mg/mL, or 200 mg/mL. In still another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the ET A R antibody provided herein is approximately 25 mg/mL, 50 mg/mL, 75 mg/mL, or 100 mg/mL.
  • the concentration of the buffer described herein is ranging approximately from 1 mM to 200 mM, from 2 mM to 50 mM, or from 5 mM to 25 mM. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the buffer described herein is approximately 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM, 40 mM, 45 mM, or 50 mM. In still another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the buffer described herein is approximately 10 mM, 15 mM, 20 mM, 25 mM, or 30 mM.
  • the buffer described herein comprises one or more selected from: citric acid, salts of citric acid, ascorbic acid, salts of ascorbic acid, gluconic acid, salts of gluconic acid, carbonic acid, salts of carbonic acid, tartaric acid, salts of tartaric acid, succinic acid, salts of succinic acid, acetic acid, salts of acetic acid, phthalic acid, salts of phthalic acid, phosphoric acid, salts of phosphoric acid, hydrochloric acid, Tris, thomethamine, and amino acids.
  • the buffer described herein is a salt of citric acid.
  • the buffer described herein is sodium citrate.
  • the buffer described herein is histidine.
  • the stable solution formulation of the ET A R antibody provided herein also comprises a surfactant.
  • the concentration of the surfactant described herein is approximately ranging from 0.001 to 1 weight/volume percent, from 0.01 to 0.5 weight/volume percent, or from 0.01 to 0.1 weight/volume percent. In another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the surfactant described herein is approximately from 0.01 to 0.1 weight/volume percent. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the surfactant described herein is approximately 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, or 0.1 weight/volume percent. In still another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the surfactant described herein is approximately 0.02, 0.03, 0.04, 0.05, or 0.06 weight/volume percent.
  • the surfactant described herein is one or more selected from sorbitan fatty acid esters, glycerin fatty acid esters, polyglycerin fatty acid esters, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene sorbitol fatty acid esters, polyoxyethylene glycerine fatty acid esters, polyethylene glycol fatty acid esters, polyoxyethylene alkyl ethers, polyoxyethylene polyoxypropylene alkyl ethers, polyoxyethylene alkylphenyl ethers, polyoxyethylene hydrogenated castor oils, polyoxyethylene beeswax derivatives, polyoxyethylene fatty acid amides, C10-C18 alkyl sulfates, polyoxyethylene C10-C16 alkyl ether sulfate with an average of 2 to 4 moles of the added oxirane groups, C1-C18 alkyl sulfosuccinate ester salts, natural surfactants, sphingophospholipids, and suc
  • the surfactant described herein is one or more selected from sorbitan fatty acid esters, e.g., sorbitan monocaprylate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan trioleate; glycerin fatty acid esters, e.g., glycerin monocaprylate, glycerin monomyristate, glycerin monostearate; polyglycerin fatty acid esters, e.g., decaglyceryl monostearate, decaglyceryl distearate, decaglyceryl monolinoleate; polyoxyethylene sorbitan fatty acid esters, e.g., polyoxyethylene sorbitan monolaurate, wherein polyoxyethylene (20) sorbitan monolaurate is TWEEN-20 and polyoxyethylene sorbitan monopalmitate is TWEEN-40, polyoxyethylene sorbitan monooleate, wherein polyoxyethylene (80), polyoxy
  • the surfactant described herein is a polyoxyethylene sorbitan fatty acid ester, e.g., TWEEN-20, TWEEN-40, TWEEN-60 and TWEEN-80. In another embodiment, the surfactant described herein is TWEEN-20 or TWEEN-80.
  • the stable pharmaceutical solution formulation of the ET A R antibody also comprises an amino acid protectant.
  • the concentration of the amino acid protectant described herein is approximately ranging from 1 mM to 500 mM or from 10 mM to 200 mM. In another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the amino acid protectant described herein is approximately from 10 mM to 200 mM. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the amino acid protectant described herein is approximately 100 mM, 110 mM, 120 mM, 130 mM, 140 mM, 150 mM, 160 mM, 170 mM, 180 mM, 190 mM, or 200 mM. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the amino acid protectant described herein is approximately 120 mM, 130 mM, 140 mM, 150 mM, or 160 mM.
  • the amino acid protectant is one or more selected from histidine, arginine, glycine, and proline. In another embodiment, the amino acid protectant described herein is one or more selected from histidine, arginine, and glycine. In yet another embodiment, the amino acid protectant described herein is arginine or a salt thereof. In still another embodiment, the amino acid protectant described herein is arginine hydrochloride.
  • the concentration of the ET A R antibody is approximately from 10 to 200 mg/mL; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the amino acid protectant is approximately from 10 to 200 mM; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, 75, or 100 mg/mL; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the amino acid protectant is approximately 140 mM; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein also comprises a polyol protectant.
  • the concentration of the polyol described herein is approximately ranging from 0.1% to 50%, from 1% to 20%, from 1% to 15%, from 2% to 10%, or from 4% to 10%. In another embodiment, in the stable pharmaceutical solution formulation of the ET A R antibody provided herein, the concentration of the polyol described herein is approximately from 4 to 10 weight/volume percent. In yet another embodiment, in the stable pharmaceutical solution formulation of the ET A R antibody provided herein, the concentration of the polyol described herein is approximately 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, or 10 weight/volume percent.
  • the polyol protectant described herein is sorbitol, mannitol, sucrose, or trehalose. In another embodiment, the polyol protectant described herein is sorbitol or mannitol.
  • the concentration of the ET A R is approximately from 10 to 200 mg/mL; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the polyol protectant is approximately from 1 to 20 weight/volume percent; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, 75 or 100 mg/mL; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the polyol protectant is approximately from 4 to 10 weight/volume percent; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the stable solution formulation of the ET A R antibody provided herein also comprises a metal chelator.
  • the concentration of the metal chelator described herein is approximately ranging from 0.001 mM to 1 mM, from 0.005 mM to 0.5 mM, or from 0.01 mM to 0.2 mM. In another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the metal chelator described herein is approximately from 0.01 mM to 0.2 mM.
  • the concentration of the metal chelator described herein is approximately 0.01 mM, 0.02 mM, 0.03 mM, 0.04 mM, 0.05 mM, 0.06 mM, 0.07 mM, 0.08 mM, 0.09 mM, or 0.1 mM. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the metal chelator described herein is approximately 0.01 mM, 0.02 mM, 0.03 mM, 0.04 mM, 0.05 mM, 0.06 mM, 0.07 mM, 0.08 mM, 0.09 mM, or 0.1 mM.
  • the concentration of the metal chelator described herein is approximately 0.03 mM, 0.04 mM, 0.05 mM, 0.06 mM, or 0.07 mM.
  • the metal chelator described herein is EDTA, DTPA, or EGTA. In another embodiment, the metal chelator described herein is EDTA.
  • the concentration of the ET A R antibody is approximately from 10 to 200 mg/mL; the concentration of the metal chelator is approximately from 0.01 to 0.2 mM; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the amino acid protectant is approximately from 10 to 200 mM; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, 75, or 100 mg/mL; the concentration of the metal chelator is approximately 0.05 mM; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the amino acid protectant is approximately 140 mM; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the concentration of the ET A R antibody is approximately from 10 to 200 mg/mL; the concentration of the metal chelator is approximately from 0.01 mM to 0.2 mM; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the polyol protectant is approximately from 1 to 20 weight/volume percent; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, or 100 mg/mL; the concentration of the metal chelator is approximately 0.05 mM; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the polyol protectant is approximately from 4 to 10 weight/volume percent; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the stable solution formulation of the ET A R antibody provided herein also comprises an antioxidant.
  • the concentration of the antioxidant described herein is approximately ranging from 0.1 mM to 50 mM, from 0.5 mM to 20 mM, or from 1 mM to 10 mM. In another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the antioxidant described herein is approximately from 1 to 10 mM. In yet another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the antioxidant described herein is approximately 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, or 10 mM. In still another embodiment, in the stable solution formulation of the ET A R antibody provided herein, the concentration of the antioxidant described herein is approximately 3 mM, 4 mM, 5 mM, 6 mM, or 7 mM.
  • the antioxidant described herein is methionine, vitamin-C, thiosulfate, thiosulfate, or benzyl methionine. In another embodiment, the antioxidant described herein is methionine.
  • the concentration of the ET A R antibody is approximately from 10 to 200 mg/mL; the concentration of the antioxidant is approximately from 1 to 10 mM; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the amino acid protectant is approximately from 10 to 200 mM; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, 75, or 100 mg/mL; the concentration of the antioxidant is approximately 5 mM; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the amino acid protectant is approximately 140 mM; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the stable solution formulation of ET A R antibody provided herein in the concentration of the ET A R antibody is approximately from 10 to 200 mg/mL; the concentration of the antioxidant is approximately from 1 mM to 10 mM; the concentration of the surfactant is approximately from 0.01 to 0.1 weight/volume percent; the concentration of the polyol protectant is approximately from 1 to 20 weight/volume percent; and the concentration of the buffer is approximately from 1 to 50 mM; wherein the pH of the formulation is approximately from 5 to 7.
  • the concentration of the ET A R antibody is approximately 25, 50, 75, or 100 mg/mL; the concentration of the antioxidant is approximately 5 mM; the concentration of the surfactant is approximately 0.04 weight/volume percent; the concentration of the polyol protectant is approximately from 4 to 10 weight/volume percent; and the concentration of the buffer is approximately 20 mM; wherein the pH of the formulation is approximately from 5 to 6.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein comprises:
  • a buffering system providing a pH of 5.0-7.0.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein comprises:
  • a buffering system providing a pH of 5.3-6.5.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein comprises:
  • a buffering system providing a pH of 5.0-7.0.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein comprises:
  • a buffering system providing a pH of 5.3-6.5.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein is an aqueous solution. In another embodiment, the stable formulation of the pharmaceutical ET A R antibody provided herein is a sterile solution.
  • the stability of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is determined by the extent of aggregation of the ET A R antibody. In one embodiment, the stable pharmaceutical solution formulation of the ET A R antibody provided herein, after stored at about 40° C. and about 75% humidity for 1, 2, 3, 6, 12, or 24 months, contains no more than 20%, 15%, 10%, 8%, 6%, 5,%, 4%, 3%, 2%, 1%, or 0.1% of an aggregated ET A R antibody.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at room temperature and about 65% humidity for 3, 6, 12, or 24 months, contains no more than 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2%, 1%, or 0.1% of an aggregated ET A R antibody.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at 2-8° C. for 6, 12, 18, 24, 36 or 48 months, contains no more than 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2%, 1%, or 0.1% of an aggregated ET A R antibody.
  • the stability of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is determined by the extent of degradation of the ET A R antibody.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at about 40° C. and about 75% humidity for 1, 2, 3, 6, 12 or 24 months, has a degree of degradation of the ET A R antibody of no more than 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2%, 1%, or 0.1%.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at room temperature and about 65% humidity for 3, 6, 12, or 24 months, has a degree of degradation of the ET A R antibody of no more than 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2%, 1%, or 0.1%.
  • the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at 2-8° C. for 6, 12, 18, 24, 36 or 48 months, has a degree of degradation of the ET A R antibody of no more than 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2%, 1%, or 0.1%.
  • the aggregation of the ET A R antibody and the loss of a monomeric ET A R antibody are determined by SEC-HPLC.
  • the stability of the stable pharmaceutical solution formulation of the ET A R antibody provided herein is determined by the change in the biological activity of the ET A R antibody.
  • the ET A R antibody in the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored at about 40° C. and about 75% humidity for 1, 2, 3, 6, 12 or 24 months, has a biological activity of no less than 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or 99.9% of its original biological activity.
  • the ET A R antibody in the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored under room temperature and about 65% humidity for 3, 6, 12, or 24 months, has a biological activity of no less than 50%, 60%, 70%, 80%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.9% of its original biological activity.
  • the ET A R antibody in the stable pharmaceutical solution formulation of the ET A R antibody provided herein after stored under temperature of 2-8° C.
  • the change in the biological activity of the ET A R antibody is determined by a calcium flux detection method to determine the ability of an ET A R antibody to inhibit an ET A R in vitro.
  • provided herein is a method of lowering hypertension (for example, PAH) in a subject, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • provided herein is a method of treating PAH in a subject, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition provided herein, for example, a stable pharmaceutical solution formulation of an ET A R antibody provided herein.
  • the term “subject” refers to a mammal, including humans, and is used interchangeably with the term “patient.”
  • treatment encompasses alleviation or prevention of at least one symptom or other aspect of a disorder, or reduction of disease severity, and the like.
  • An antibody provided herein needs not to provide a complete cure, or to eradicate every symptom or manifestation of a disease, to be an effective therapeutic agent.
  • therapeutic agents can reduce the severity of a given disease state, but need not to abolish every manifestation of the disease to be effective.
  • a prophylactic agent needs not to prevent the onset of a condition completely in order to be effective.
  • One embodiment of the invention is directed to a method comprising administering to a patient an antibody in an amount and for a time sufficient to induce a sustained improvement over baseline of an indicator that reflects the severity of a particular disorder.
  • a pharmaceutical composition can be administered by any suitable technique, including, but not limited to, parenterally, topically, or by inhalation. If injected, the pharmaceutical composition can be administered, for example, via an intra-articular, intravenous, intramuscular, intralesional, intraperitoneal or subcutaneous route, by bolus injection or continuous infusion. It is considered, for example, localized administration at the disease or injury site, such as transdermal administration and sustained release of an implant. Delivery by inhalation includes, for example, nasal or oral inhalation, use of a nebulizer, inhalation of an antibody in aerosol form, and the like. Other alternatives include oral preparations, including pills, syrups, or lozenges.
  • the antibodies provided herein are administered in a composition comprising one or more additional components such as a physiologically acceptable carrier, excipient or diluent.
  • the composition additionally comprises one or more physiologically active agents as described below.
  • the composition comprises one, two, three, four, five, or six physiologically active agents in addition to one or more antibodies (e.g., murine antibodies or humanized antibodies) provided herein.
  • the pharmaceutical composition comprises a murine antibody or humanized antibody of the invention together with one or more substances selected from the group consisting of a buffer suitable for the antibody at a suitable pH, an antioxidant such as ascorbic acid, a low molecular weight polypeptide (such as those having fewer than 10 amino acids), a protein, an amino acid, a carbohydrate such as dextrin, a chelating agent such as EDTA, glutathione, a stabilizer, and an excipient.
  • preservatives can also be added.
  • the composition can be formulated as a lyophilizate using appropriate excipient solutions as diluents. Suitable components are nontoxic to recipients at the dosages and concentrations employed.
  • kits for use by medical practitioners are provided, including one or more antibodies of the invention and a label or other instructions for use in treating any of the conditions discussed herein.
  • the kit includes a sterile preparation of one or more human antibodies, which can be in the form of a composition as disclosed above, and can be in one or more vials.
  • Dosages and the frequency of administration can vary according to such factors as the route of administration, the particular antibodies employed, the nature and severity of the disease to be treated, whether the condition is acute or chronic, and the size and general condition of the subject. Appropriate dosages can be determined by procedures known in the pertinent art, e.g. in clinical trials that can involve dose escalation studies.
  • An antibody provided here can be administered, for example, once or more than once, e.g., at regular intervals over a period of time.
  • a murine antibody or humanized antibody is administered over a period of at least once a month or more, e.g., for one, two, or three months or even indefinitely.
  • long-term treatment is generally most effective.
  • administration for shorter periods, e.g., from one to six weeks, can be sufficient.
  • the humanized antibody is administered until the patient manifests a medically relevant degree of improvement over baseline for the chosen indicator or indicators.
  • therapeutic regimens comprise subcutaneous injection of an antibody once a week, at an appropriate dosage, to treat a condition in which pulmonary arterial pressure levels play a role. Weekly or monthly administration of antibody would be continued until a desired result is achieved, e.g., the subject's symptoms subside. Treatment can resume as needed, or, alternatively, maintenance doses can be administered.
  • a subject's levels of pulmonary arterial pressure can be monitored before, during and/or after treatment with an antibody such as a humanized antibody, to detect changes, if any, in their levels.
  • an antibody such as a humanized antibody
  • the incidence of elevated pulmonary arterial pressure can vary according to such factors as the stage of the disease.
  • Known techniques can be employed for measuring pulmonary arterial pressure levels.
  • compositions of the invention involve the use of an antibody and one or more ET A R antagonists for example, two or more antibodies of the invention, or an antibody of the disclosure and one or more other ET A R antagonists.
  • an antibody is administered alone or in combination with other agents useful for treating the condition with which the patient is afflicted. Examples of such agents include both proteinaceous and non-proteinaceous drugs.
  • co-administration and combination therapy are not limited to simultaneous administration, but also include treatment regimens in which an antibody is administered at least once during a course of treatment that involves administering at least one other therapeutic agent to the patient.
  • the method of preparing a medicament for treating pulmonary arterial hypertension and related disorders comprises a mixture of the antibody provided herein and pharmaceutically acceptable excipients.
  • the preparation method of the medicament was as described above.
  • a composition, kit, and method related to an antibody specifically binding to a human endothelin receptor are further provided herein.
  • Nucleic acid molecules and derivatives and fragments thereof comprising a part of or a full polynucleotide encoding a polypeptide interacting with an ET A R, for example, nucleic acids encoding all or part of an endothelin receptor antibody, an antibody fragment or an antibody derivative, are also provided.
  • a vector and plasmid comprising nucleic acids and cells and cell line comprising nucleic acids and/or a vector and plasmid are further provided herein.
  • Methods provided herein include, for example, methods for preparation, identification or separation of an antibody interacting with a human ET A R, for example, a method of an ET A R antibody, a method for determining whether an antibody binds to ET A R, and a method for administering an antibody binding to an ET A R to an animal model.
  • CHO-DHFR-cells were seeded into a 6-well plate. After 24 h culture, the cells were transfected with a pIRES plasmid (Clontech, commercial) modified to carry hET A R gene (see SEQ ID NO: 1 for the nucleotide sequence, and SEQ ID NO: 2 for the amino acid sequence). The transfection was carried out by following the transfection conditions recommended by Invitrogen for Lipofectamine 2000. Forty-eight hours after transfection, the medium was replaced with a complete medium containing 10 nM MTX (methotrexate). The medium was changed every 3 days for about two weeks until stable clones appeared. The dispersed cell colonies were detached from the plate and collected.
  • pIRES plasmid (Clontech, commercial) modified to carry hET A R gene (see SEQ ID NO: 1 for the nucleotide sequence, and SEQ ID NO: 2 for the amino acid sequence). The transfection was carried out by following the transfection conditions recommended by Invit
  • mice An emulsion of the CHO-DHFR-hET A R whole cells and Freund's adjuvant was injected subcutaneously into BALB/c mice (6-8 weeks) at 2 ⁇ 10 6 cells/mouse. After 2 weeks, the mice were boosted with incomplete Freund's adjuvant emulsified immunogen and then boosted once every week. After immunization for 6 times in total, blood samples were collected from the clipped tail ends and centrifuged to collect the serum. The serum was analyzed for serum titers by FACS. After the acceptable antibody titers were achieved, the mice were sacrificed and their spleen cells were harvested under aseptic conditions. SP2/0 cells were collected at the logarithmic phase of growth with 3 min centrifugation at 2,000 rpm.
  • the cell pellets were resuspended with serum-free culture medium, then centrifuged, resuspended for a second time and counted.
  • Spleen cells and SP2/0 cells were mixed at ratio of SP2/0 cells:spleen cells ⁇ 1:1, followed by 3 rounds of washing-centrifugation.
  • 1 mL of pre-warmed PEG-1350 was added dropwise (finished in 30 s), after pipette-mixing for 1 min, 30 mL of the pre-warmed serum-free medium (Invitrogen) was added slowly to terminate the PEG fusion.
  • the cell pellets were resuspended in the fusion culture medium.
  • Spleen cells (20,000) and feeder layer cells (5,000) in 100 ⁇ L were plated into each well of 96-well plates.
  • Fused hybridoma cells and feeder layer cells were co-cultured in 96-well plates with HAT (sarcine, amethopterin and thymidine) selection to get rid of the non-fused cells.
  • HAT sarcine, amethopterin and thymidine
  • CHO-DHFR-hET A R cells over-expressing hET A R and CHO-DHFR-cells not expressing hET A R were separately transferred into a 96-well plate and allowed to grow to 90% confluent. The supernatant of the culture medium was removed and attached cells were washed twice with PBS, then 100 ⁇ L, 100% methanol was added to fix the cells for 10 min at 4° C. Then 100 ⁇ L freshly made 0.6% H 2 O 2 -PBS was added, and after incubation at room temperature for 20 min, the cells were washed twice with PBS. After blocked with PBS-1% BSA solution, the hybridoma supernatant was added and incubated for 90 min at 4° C.
  • Hybridoma cells secreting antibodies were collected.
  • Hybridoma mRNA was extracted according to the manufacturer protocol of QIAGEN mRNA extraction kit. Then the extracted mRNA was transcribed reversely into cDNA.
  • the reverse transcription primers were specific primers for the light and heavy chain constant regions of a mouse, with the heavy chain reverse transcription primer being (5′-TTTGGRGGGAAGATGAAGAC-3′) (SEQ ID NO: 199), the light chain reverse transcription primers being (5′-TTAACACTCTCCCCTGTTGAA-3′) (SEQ ID NO: 200) and (5′-TTAACACTCATTCCTGTTGAA-3′) (SEQ ID NO: 201).
  • RT-PCR reaction conditions were as following: 25° C. for 5 min, 50° C.
  • Reversely transcribed cDNA was diluted with 0.1 mM TE to 500 ⁇ L, added into the ultrafiltration centrifuge tube (Amicon Ultra-0.5) and centrifuged at 2,000 g for 10 min. The filtrate was removed, 500 ⁇ L of 0.1 mM TE were added and centrifuged at 2,000 g for 10 min. The filtrate was removed and the preparation tube was placed in inversion to the new centrifugal tube, and centrifuged at 2,000 g for 10 min to obtain the purified cDNA.
  • Purified cDNA (10 ⁇ L) was taken as a template, followed by addition of 4 ⁇ L 5 ⁇ tailing buffer (Promega), 4 ⁇ L dATP (1 mM) and 10 U terminal transferase (Promega), mixing uniformly, and incubation at 37° C. for 5 min and then at 65° C. for 5 min.
  • the PolyA tail cDNA was used as a template and PCR was performed to amplify light and heavy chain variable region genes of antibodies.
  • Upstream primers were all oligodT, with heavy chain downstream primers being (5′-TGGACAGGGATCCAGAGTTCC-3′) (SEQ ID NO: 202) and (5′-TGGACAGGGCTCCATAGTTCC-3′) (SEQ ID NO: 203), and light chain downstream primer being (5′-ACTCGTCCTTGGTCAACGTG-3′) (SEQ ID NO: 204).
  • the PCR reaction conditions were: 95° C. for 5 min; 95° C. for 30 s, 56° C. for 30 s, 72° C. for 1 min, 40 cycles; and 72° C. for 7 min.
  • the PCR products were connected to the PMD 18-T vector (Takara Bio) for sequencing.
  • the sequences of the antibody clones were listed in Table 2.
  • PCR primers were designed based on the DNA sequences of the antibodies, thus the complete light chain, heavy chain signal peptides and variable domains and mouse IgG1 constant region were ligated into expression vector pTM5.
  • sequences of light and heavy chain variable regions of the screened mouse antibodies were aligned with the homologous antibodies, using NCBI online antibody variable region sequence alignment tool (Ig Blast) to search the germline gene sequences of a humanized antibody (Ig Germline Gene sequence) homologous to the selected antibodies variable region sequence for humanization, and the humanized gene sequence with highest homology except CDR sequences was used as a template for CDR grafting to obtain humanized antibody variable region sequences and to synthesize humanized antibody light and heavy chain genes through a CRO.
  • NCBI online antibody variable region sequence alignment tool Ig Blast
  • PCR primers were designed and appropriate restriction enzyme sites were introduced at the 5′ ends and 3′ ends.
  • the humanized antibody variable regions were amplified and then combined with the human IgG2 or IgG4 constant region sequence to obtain whole recombinant humanized antibody sequences.
  • the expression of the recombinant antibodies was achieved according to step 7, and their affinities to ET A R was analyzed by FACS as described in step 9.
  • the best humanized antibody candidate retaining affinity to ET A R was selected from the group, and its variable region sequence was further improved by site-specific mutagenesis for improved affinity to ET A R.
  • the heavy and light chain variable region gene sequences of an optimized humanized antibody were synthesized by Genscript Biotechnology CO., LTD by introducing two restriction sites of NheI at the 5′-end and SalI at the 3′-end.
  • the whole heavy chain variable region was ligated with a heavy chain constant region in an expression vector of pTM5.
  • the light chain variable region was ligated with a light chain constant region in the expression vector of pTM5.
  • a suspension of an HEK293 or CHO expressing cell line (5 ⁇ 10 5 /mL) was inoculated to a shaker flask. After 24 h rotation at 37° C., the cell density reached 1 ⁇ 10 6 /mL and were ready for transfection.
  • Polyethylenimine (PEI) was used as a transfection reagent with an optimal mixing ratio of 3:1 for PEI to DNA (DNA amount, 0.5 ⁇ g/L ⁇ 10 6 cells; the ratio of the antibody light chain DNA and antibody heavy chain DNA, 3:2).
  • a mixture of both was added into the cell culture after 15 min incubation.
  • the cells after treated with the PEI/DNA mixture were rotated for more than 24 h at 37° C. and 5% CO 2 . Then 0.5% of tryptone was added into the cell culture as a supplement required by expression, and after the completion of expression (more than 96 h), the cell supernatant was collected for the antibody purification and separation.
  • PBS containing 10 mM EDTA was used to detach and collect 10 5 CHO-DHFR-hET A R cells into a 1.5 mL EP tube. The supernatant was removed after centrifugation and the negative control sample was resuspended with a loading buffer (PBS, 2% FBS). For the positive control, 200 ⁇ L antibody supernatant was added to resuspension cells and incubation at room temperature; the cells were then centrifuged at 1500 rpm to remove the supernatant, washed with a FACS loading buffer and centrifuged again.
  • PBS 2% FBS
  • the cells were resuspended with addition (200 ⁇ L/well) of a FITC labeled goat anti-mouse fluorescent antibody at 1:50 dilution (BD Pharmingen) and incubated at room temperature for 30 min in the dark. Supernatant was removed after centrifugation, cells were washed with FACS loading buffer, centrifuged again and resuspended with the loading buffer for analysis. The recombinant antibody supernatant and CHO-DHFR-hET A R cells had specific binding. Gray peak and dotted line peak were negative controls; the solid line peak, corresponding to the antibody supernatant, moved to the right significantly.
  • CHO-DHFR cells co-expressing hET A R-Aequorin were seeded into a 96-well cell culture plate with 25000 cells per well and cultured at 37° C. overnight. The next day the culture supernatant was removed. Coelenterazine (50 ⁇ L) (Promega) was added in the dark and incubated at 37° C. for 2 h, and then 50 ⁇ L of a hybridoma supernatant or a purified antibody were added and incubated at 37° C. for 30 min. After the incubation, 50 ⁇ L endothelin 1 was added and the changes of calcium influx within 40 s were recorded by a SpectraMax L microplate reader (Molecular Devices).
  • the acute hypoxia-induced pulmonary arterial hypertension (PAH) model of cynomolgus was codeveloped with Crown Bioscience Inc. (Taicang), and the efficacy of A-1 antibody as a single intravenous injection was evaluated in this PAH model. All animals were fasted overnight and weighed, and then received a single intravenous injection of 10 mg/kg of A-1 antibody. Three hours later, the animals were anesthetized. The tricuspid regurgitation velocity by Doppler color echocardiography along with heart rate and oxygen saturation were monitored simultaneously.
  • the baseline was obtained and the induction of 12% hypoxia was followed and at the same time the tricuspid regurgitation velocity was measured; Analysis was made to determine if the antibody would improve hypoxia-induced pulmonary arterial pressure under 12% hypoxia.
  • the tests were performed again.
  • the animals were anesthetized, the tricuspid regurgitation velocity by Doppler color echocardiography along with heart rate and oxygen saturation were monitored simultaneously.
  • the baseline was obtained and the induction of 12% hypoxia was followed and at the same time the tricuspid regurgitation velocity was measured. Analysis was made to determine if the antibody would still improve hypoxia-induced pulmonary arterial pressure. If the efficacy maintained after 48 h, 96 h later, hypoxia induction experiment was performed again.
  • the area under the curve of pulmonary artery systolic pressure versus time was calculated, and by comparing the area under the curve, it was found that A-1 maintained the efficacy of reducing pulmonary artery pressure within 96 h.
  • CE-SDS Capillary Electrophoresis
  • a capillary electrophoresis apparatus (Beckmann MDQP/ACE) was used, and samples were treated with an IgG purity/heterogeneity assay kit (Beckmann) by adding beta-mercaptoethanol to reduced samples or iodoacetamide to non-reduced samples. Beckmann non-coated capillaries were used to separate, and the samples were loaded at a concentration of 1 mg/mL automatically. After finishing loading a sample, separation was performed at 15 kV reverse voltage, and a UV214 nm absorbance time curve was recorded. When the process was finished, the absorption peaks at UV214 nm of the main peak, fragments and aggregates were integrated.
  • the ratio of the main peak over total area was calculated, which represents the purity of the non-reduced sample.
  • the ratio of light and heavy chain peak areas over the total area was calculated, which represents the purity of the reduced sample.
  • Reference antibody A-1 and test samples were diluted serially, and 50 ⁇ L/well of the dilutions were added to the 96 well plate (in the dark). After addition of the samples, the plate was placed in an incubator at 37° C. and 5% CO 2 for another 30 min. The fluorescence intensity of each well was read on a microplate reader. The read values of three wells with only DMEM/F12 medium (no phenol red) were the background values, and the rest of the wells were added with 20 nM endothelin-1.
  • the read data were pasted to Excel for analysis, and the time point of the peak fluorescent intensity was selected as the calculation value.
  • the average of the fluorescence values of the blank controls was obtained, and the average value was then substracted from each original value.
  • the values of the maximum fluorescence intensities were averaged, and the relative percentage of each well to the average of the maximum response average was calculated finally.
  • the percentage and concentration of each well were used to calculate IC 50 values for the reference ET A R antibody and the test samples, as well as curve fitting correlation coefficients R 2 .
  • the biological activity of a test sample (%) (IC 50 of the reference/IC 50 of the test sample) ⁇ 100.
  • the experimental conditions were freeze-thaw: freeze at ⁇ 20° C., thaw at room temperature, 3 cycles; high temperature: 37° C. for 10 days, 13 days; illumination: 5000 lx, 300 ⁇ W/cm 2 , 25° C., 5 days; testing parameters: appearance, visible particles, purity (SEC-HPLC, non-reduced CE-SDS), charge variant.
  • the second group of experiments were performed based on the first group of experiments, and sodium citrate was selected as the buffer system.
  • the concentration of sodium citrate was 20 mM, pH was 5.8, and the concentration of antibody A-1 was about 30 mg/mL.
  • arginine was replaced with arginine hydrochloride, and its concentration was 140 mM.
  • the experiment for the selection of a protectant was performed, and conditions of high temperature and illumination were modified to 40° C./2 watts, 1 month, 5000 lx, 0 day, 2 days, 5 days, 10 days.
  • the design plan is shown in Table 10.
  • the sodium citrate was used as the buffer, and the results indicates that, after high temperature and illumination, the formulation of antibody A-1 showed slight opalescence (see Table 14). Comparing the effects of several protectants, we considered arginine hydrochloride the best in preserving the purity of antibody A-1 (see Table 15). The illumination induced significant change of charge variants (see Table 16), therefore, it was recommended to store antibody A-1 in the dark.
  • the experiment was to evaluate the protective effect of 0.0187 mg/mL EDTA, and two buffers were selected, 20 mM histidine salt and 20 mM sodium citrate with 0.02% and 0.1% of Tween-80 (Table 17).
  • the concentration of antibody A-1 was 60 mg/mL
  • arginine hydrochloride was 140 mM
  • pH was 5.8.
  • the three formulation samples were subject to accelerated degradation experiment, and the experimental condition was illumination: 5000 lx, 300 ⁇ W/cm 2 , 25° C., 3 days, 6 days; the test items: appearance, visible particles, purity (SEC-HPLC), and charge variant.
  • methionine was added as a screening antioxidant, and the detailed plan was in Table 18.
  • the experiment conditions were: high temperature 40° C. for 2 weeks, 1 month; illumination 5000 lx for 0 day, 2 days, 5 days, 10 days.
  • Methionine formulation screening table Arginine For- Con- Sodium hydrochloride mu- centration citrate (%) Methionine Tween- lation (mg/mL) (mM) (mM) (mM) 80 pH F1-M ⁇ 50 20 140 — 0.04 5.8 F2-M ⁇ 50 20 140 5 0.04 5.8 F3-M ⁇ 50 20 140 10 0.04 5.8
  • antibody A-1 (20 mM sodium citrate, 140 mM arginine hydrochloride, 0.04% TWEEN-80, pH 5.8) was tested at 3 different antibody concentrations in long-term and accelerated stability studies. After 9 months, regardless of acceleration and long-term stability studies, the quality of the protein still met the quality standards set. Especially after 9 months of long-term storage, the purity of antibody A-1 remained above 98%.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Endocrinology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Dermatology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
US16/305,828 2016-05-31 2017-05-27 Etar antibody, and pharmaceutical compositions and use thereof Abandoned US20190134196A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201610376600 2016-05-31
CN201610376600.7 2016-05-31
CN201610954533.2 2016-10-27
CN201610954533.2A CN107987162A (zh) 2016-10-27 2016-10-27 Etar抗体,其药物组合物及其应用
PCT/CN2017/086369 WO2017206840A1 (zh) 2016-05-31 2017-05-27 Etar抗体,其药物组合物及其应用

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/086369 A-371-Of-International WO2017206840A1 (zh) 2016-05-31 2017-05-27 Etar抗体,其药物组合物及其应用

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/155,644 Continuation US20230346932A1 (en) 2016-05-31 2023-01-17 Etar antibody, and pharmaceutical compositions and use thereof

Publications (1)

Publication Number Publication Date
US20190134196A1 true US20190134196A1 (en) 2019-05-09

Family

ID=60479108

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/305,828 Abandoned US20190134196A1 (en) 2016-05-31 2017-05-27 Etar antibody, and pharmaceutical compositions and use thereof
US18/155,644 Pending US20230346932A1 (en) 2016-05-31 2023-01-17 Etar antibody, and pharmaceutical compositions and use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/155,644 Pending US20230346932A1 (en) 2016-05-31 2023-01-17 Etar antibody, and pharmaceutical compositions and use thereof

Country Status (8)

Country Link
US (2) US20190134196A1 (ja)
EP (1) EP3476864A4 (ja)
JP (1) JP7090557B2 (ja)
KR (1) KR102549008B1 (ja)
AU (1) AU2017273959A1 (ja)
CA (1) CA3025714A1 (ja)
TW (1) TWI826351B (ja)
WO (1) WO2017206840A1 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105669863B (zh) 2014-12-05 2019-09-13 鸿运华宁(杭州)生物医药有限公司 一种能与人内皮素受体特异性结合的抗体及其应用
FR3077574B1 (fr) * 2018-02-07 2022-04-01 Commissariat Energie Atomique Anticorps dirige contre le sous-type a des recepteurs aux endothelines et ses utilisations.

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090304706A1 (en) * 2006-04-21 2009-12-10 Novartis Ag Antagonist anti-cd40 antibody pharmaceutical compositions

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
DE3675588D1 (de) 1985-06-19 1990-12-20 Ajinomoto Kk Haemoglobin, das an ein poly(alkenylenoxid) gebunden ist.
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5011912A (en) 1986-12-19 1991-04-30 Immunex Corporation Hybridoma and monoclonal antibody for use in an immunoaffinity purification system
US5262522A (en) 1991-11-22 1993-11-16 Immunex Corporation Receptor for oncostatin M and leukemia inhibitory factor
CA2146559A1 (en) 1992-10-23 1994-05-11 Melanie K. Spriggs Methods of preparing soluble, oligomeric proteins
US5457035A (en) 1993-07-23 1995-10-10 Immunex Corporation Cytokine which is a ligand for OX40
US6133426A (en) 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
ES2301198T3 (es) 1997-06-12 2008-06-16 Novartis International Pharmaceutical Ltd. Polipeptidos artificiales de anticuerpos.
GB9722131D0 (en) 1997-10-20 1997-12-17 Medical Res Council Method
US6210924B1 (en) 1998-08-11 2001-04-03 Amgen Inc. Overexpressing cyclin D 1 in a eukaryotic cell line
GB9928787D0 (en) 1999-12-03 2000-02-02 Medical Res Council Direct screening method
US20020055457A1 (en) * 2000-08-07 2002-05-09 Janus Todd J. Methods of treating cancer and the pain associated therewith using endothelin antagonists
GB0025144D0 (en) 2000-10-13 2000-11-29 Medical Res Council Concatenated nucleic acid sequences
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20040202995A1 (en) 2003-04-09 2004-10-14 Domantis Nucleic acids, proteins, and screening methods
DOP2006000029A (es) * 2005-02-07 2006-08-15 Genentech Inc Antibody variants and uses thereof. (variantes de un anticuerpo y usos de las mismas)
JP2008532519A (ja) * 2005-03-09 2008-08-21 ボード オブ リージェンツ, ザ ユニバーシティ オブ テキサス システム 遺伝子発現のインビボ画像化に関する方法および組成物
CN101426527A (zh) * 2006-02-03 2009-05-06 米迪缪尼有限公司 蛋白质制剂
JP2008280266A (ja) * 2007-05-09 2008-11-20 Sekisui Chem Co Ltd モノクローナル抗体
PE20091174A1 (es) * 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd Formulacion liquida con contenido de alta concentracion de anticuerpo
JP2010006745A (ja) * 2008-06-27 2010-01-14 Sekisui Chem Co Ltd 融合タンパク質、融合タンパク質固定化担体、化合物のスクリーニング方法、スクリーニング用組成物、並びに、スクリーニング用キット
JP2010138165A (ja) * 2008-11-14 2010-06-24 Sekisui Chem Co Ltd 抗ヒトエンドセリン受容体タイプaモノクローナル抗体、並びに、ハイブリドーマ
WO2011109365A2 (en) * 2010-03-01 2011-09-09 Progenics Pharmaceuticals, Inc. Concentrated protein formulations and uses thereof
WO2012076670A2 (en) * 2010-12-10 2012-06-14 Novartis Ag Antibody formulation
US20140004131A1 (en) * 2012-05-04 2014-01-02 Novartis Ag Antibody formulation
CN103728454B (zh) * 2012-10-10 2016-05-18 吴庄民 基于表位抗原肽的抗内皮素受体a抗体酶联免疫试剂盒及其应用
CN104513313B (zh) * 2013-09-27 2018-11-09 鸿运华宁(杭州)生物医药有限公司 一种跨膜受体的单克隆抗体的筛选方法
CN105669863B (zh) * 2014-12-05 2019-09-13 鸿运华宁(杭州)生物医药有限公司 一种能与人内皮素受体特异性结合的抗体及其应用

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090304706A1 (en) * 2006-04-21 2009-12-10 Novartis Ag Antagonist anti-cd40 antibody pharmaceutical compositions

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation

Also Published As

Publication number Publication date
US20230346932A1 (en) 2023-11-02
JP7090557B2 (ja) 2022-06-24
TWI826351B (zh) 2023-12-21
AU2017273959A1 (en) 2019-01-03
KR20230098919A (ko) 2023-07-04
EP3476864A4 (en) 2020-02-12
WO2017206840A1 (zh) 2017-12-07
KR20190013918A (ko) 2019-02-11
EP3476864A1 (en) 2019-05-01
CA3025714A1 (en) 2017-12-07
KR102549008B1 (ko) 2023-06-30
JP2019523764A (ja) 2019-08-29
TW201805023A (zh) 2018-02-16

Similar Documents

Publication Publication Date Title
US11712470B2 (en) Human endothelin receptor antibody and use thereof
US20230346932A1 (en) Etar antibody, and pharmaceutical compositions and use thereof
US20220411520A1 (en) Gipr antibody and fusion protein between same and glp-1, and pharmaceutical composition and application thereof
US11780916B2 (en) GIPR antibody and GLP-1 fusion protein thereof, and pharmaceutical composition and application thereof
US20220265771A1 (en) Fusion protein of eta antibody and tgf-beta trap and pharmaceutical composition and application thereof
US11542336B2 (en) GCGR antibody and GLP-1 fusion protein thereof, pharmaceutical composition thereof and application thereof
US20210163614A1 (en) Apj antibody, fusion protein thereof with elabela, and pharmaceutical compositions and use thereof
KR102669444B1 (ko) Etar 항체 및 이의 약학 조성물 및 용도
RU2800370C2 (ru) Антитело к gipr и его слитый с glp-1 белок, а также фармацевтическая композиция на его основе и его применение
JP2024514253A (ja) ヒトエンドセリン受容体に特異的に結合できる抗体及び糖尿病性腎症及び慢性腎疾患の治療におけるその応用

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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