HK1148230A - Methods of treating inflammatory pain - Google Patents
Methods of treating inflammatory pain Download PDFInfo
- Publication number
- HK1148230A HK1148230A HK11102514.0A HK11102514A HK1148230A HK 1148230 A HK1148230 A HK 1148230A HK 11102514 A HK11102514 A HK 11102514A HK 1148230 A HK1148230 A HK 1148230A
- Authority
- HK
- Hong Kong
- Prior art keywords
- cgrp
- antibody
- seq
- pain
- inflammatory pain
- Prior art date
Links
Description
Technical Field
The present invention relates to anti-CGRP antibodies for use in the prevention and/or treatment of inflammatory pain and/or symptoms of inflammatory pain, and to methods of treating and/or preventing inflammatory pain and/or symptoms of inflammatory pain using anti-CGRP antibodies.
Background
Inflammatory processes are a complex series of biochemical and cellular events that are activated in response to tissue damage or the presence of foreign substances, which lead to swelling and Pain (Levine and Taiwo, 1994, Textbook of Pain, 45-56). Arthritic pain is the most common inflammatory pain. Rheumatoid disease is one of the most common chronic inflammatory conditions in developed countries and rheumatoid arthritis is a common cause of disability (disability). The exact etiology of rheumatoid arthritis is not clear, but current assumptions suggest that both genetic and microbial factors may be important (Grennan & Jayson, 1994, Textbook of Pain, 397-. It is estimated that approximately 1600 million Americans suffer from symptomatic Osteoarthritis (OA) or degenerative joint disease, most of them over the age of 60 years, and this value is expected to increase to 4 million with increasing population age, making this a huge public health problem (Houge & Mersfelder, 2002, Ann Pharmacother., 36, 679-. Most patients with osteoarthritis seek medical attention due to the attendant pain. Arthritis has a major impact on psychosocial and physical functioning and is known to be the leading cause of disability in the later years. Ankylosing spondylitis is also a rheumatic disease that causes arthritis of the spine and sacroiliac joints. It varies from lifelong intermittent episodes of back pain to severe chronic diseases that attack the spine, surrounding joints and other body organs.
Another type of inflammatory pain is visceral pain, which includes pain associated with Inflammatory Bowel Disease (IBD). Visceral pain is pain associated with the viscera (which includes the organs of the abdominal cavity). These organs include the sexual organs, spleen and parts of the digestive system. Visceral associated pain can be divided into digestive visceral pain and non-digestive visceral pain. Commonly encountered Gastrointestinal (GI) disorders that cause pain include Functional Bowel Disorders (FBD) and Inflammatory Bowel Disease (IBD). Such GI disorders include a wide range of disease states that are currently only moderately controlled, including gastroesophageal reflux, dyspepsia, Irritable Bowel Syndrome (IBS) and Functional Abdominal Pain Syndrome (FAPS) for FBD and Crohn's disease, ileitis and ulcerative colitis for IBD, all of which frequently produce visceral pain. Other types of visceral pain include pain associated with dysmenorrhea, cystitis and pancreatitis, and pelvic pain.
There is an urgent medical need to identify novel pharmaceutically active compounds which interfere with the key steps of the inflammatory pain process and which are particularly useful for the treatment and/or prevention of arthritic pain and/or the symptoms of arthritic pain.
Surprisingly, we have found that administration of an anti-CGRP antibody is effective in the prevention and/or treatment of inflammatory pain, arthritic pain, in particular osteoarthritic pain.
CGRP (calcitonin gene-related peptide) is a 37 amino acid neuropeptide used as a neurotransmitter in the central nervous system. It binds with high affinity to the CGRP receptor, calcitonin receptor-like receptor (CRLR), thereby activating adenylate cyclase and protein kinase a production.
Small molecule selective CGRP antagonists administered by central penetrating the spine have been shown to be useful in the treatment of neuropathic and nociceptive Pain conditions (Adwanikar et al, Pain2007), suggesting that removal of endogenous CGRP in the spinal cord has an analgesic effect. Furthermore, intrathecal administration of anti-CGRP antisera has been shown to reduce nociceptive behaviour in rodent models of arthritis (Kuraishi, y., et al neurosci. lett (1998)92, 325-329).
Surprisingly, we have found that administration of an anti-CGRP antibody, when administered peripherally, is effective in the prevention and/or treatment of inflammatory pain, particularly osteoarthritis pain, for the site of peripheral action. This peripheral route of administration offers significant advantages over the need for intrathecal or spinal administration of antibodies (a more dangerous and inconvenient procedure).
Summary of The Invention
The present invention provides the use of an anti-CGRP antagonist antibody for the manufacture of a medicament for the prevention and/or treatment of inflammatory pain and/or symptoms of inflammatory pain, wherein the medicament is prepared for peripheral administration.
The invention also provides a method of preventing and/or treating inflammatory pain and/or symptoms of inflammatory pain in an individual comprising peripherally administering to the individual a therapeutically effective amount of an anti-CGRP antagonist antibody.
In one embodiment, the anti-CGRP antagonist antibody acts peripherally after administration.
Brief description of the drawings
FIG. 1: a pain model for osteoarthritis. Antibody G2 was administered intravenously (1ml/kg, IV) at 1 and 10mg/kg and null antibodies (which did not bind CGRP) at 10mg/kg, IV as negative controls. Both antibodies were dissolved in vehicle solution containing PBS + 0.01% Tween 20. Celecoxib (Celecoxib) was used as a positive control in this study. It was suspended in 0.5% methylcellulose and 0.025% Tween-20 and administered at 30mg/kg by oral feeding (1ml/kg) 2 times per day throughout the study period. Pain responses were assessed on days 2, 3, 7 and 10 after the start of the pharmacological study (day 0), in a totally blind manner. Data are mean ± SEM of 6 rats per group. P < 0.05 and p < 0.01, compared to baseline values (Dunnett's test in GraphPad Prism). In the figure, from left to right, bar 1 is baseline, bars 2-5 are null antibodies, bars 6-9 are 1mg/ml G2, bars 10-13 are 10mg/ml G2, and bars 14-17 are celecoxib 30 mg/ml.
FIG. 2: binding assay data demonstrating that antibody G1 inhibits the binding of α -CGRP to CGRP1 receptor.
FIG. 3 a: serum levels of anti-CGRP concentration (ug/ml) measured by anti-IgG ELISA versus time after IV administration of 10 mg/kg.
FIG. 3 b: serum levels of anti-CGRP concentration (ug/ml) measured by anti-IgG ELISA versus time after IV administration of 10, 30, 100 mg/kg.
FIG. 4: alanine scanning using the C-terminal CGRP fragment (CGRP 25-37). The change in affinity is expressed as fold loss of affinity, and it shows that anti-CGRP antibody G1 binds to the C-terminal region of human α -CGRP.
FIG. 5: solution competition with Biacore: CGRP, CGRP fragments or peptides related in sequence to CGRP are used to determine the specificity of G1.
FIG. 6: CGRP sequences from human, cynomolgus monkey, rat, mouse, dog and rabbit. Non-conserved residues between species are underlined and the epitope for G1 is in bold.
FIG. 7: data showing that G1 inhibited neurogenic flare (neurogenic flare) in the skin (starting 90 minutes after treatment). G1(1ml/kg) was administered by intravenous administration. Data were from 6-8 or 13 rats per group. P-0.05, p-0.01, compared to vehicle (phosphate buffered saline) treatment group at each time point (AVOVA).
Table 1: kd of anti-CGRP antibodies against human α -CGRP and IC50[ muMab7E9 ═ murine precursor of G1, measured at 25 ℃. Its K for rat beta-CGRPD1 nM. RN4901 ═ murine tool (murine tool), which recognizes the same epitope as G1 but shows the same affinity and selectivity in rats (β -CGRP K)D17 nM); g1 ═ antibody humanized from muMab7E9 (K for rat β CGRP)D=0.1nM)]。
Table 2: g1 binding affinity measured using Biacore.
Description of the invention
General techniques
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the capabilities of a person of ordinary skill in the art. Such techniques are explained extensively in literature, for example, Molecular Cloning: a Laboratory Manual, 2 nd edition (Sambrook et al, 1989) Cold Spring Harbor Press; oligonucletoideosynthesis (m.j.gait, ed., 1984); method in Molecular Biology, Humana Press; cell Biology: a Laboratory Notebook (J.E. Cellis, ed., 1998) Academic Press; animal Cell Culture (r.i. freshney, ed., 1987); introduction to Cell and Tissue Culture (J.P.Mather and P.E.Roberts, 1998) Plenum Press; cell and Tissue Culture: laboratory Procedures (a.doyle, j.b.griffiths, and d.g.newell, eds., 1993-1998) j.wiley and Sons; methods in Enzymology (academic Press, Inc.); handbook of Experimental Immunology (d.m.weir and c.c.blackwell, eds.); gene Transfer Vectors for Mammalian Cells (j.m.miller and m.p.calos, eds., 1987); current Protocols in molecular Biology (F.M. Ausubel et al, eds., 1987); and (3) PCR: the polymerase Chain Reaction, (Mullis et al, eds., 1994); CurrentProtocols in Immunology (j.e. coligan et al, eds., 1991); short protocols in Molecular Biology (Wiley and Sons, 1999); immunobiology (c.a. janeway and p.travers, 1997); antibodies (p.finch, 1997); antibodies: a practical proproach (D.Catty., ed., IRL Press, 1988-; monoclone antigens: a practical approach (P.shepherd and C.dean, eds., Oxford University Press, 2000); usingentibodes: a Laboratory manual (E.Harlow and D.Lane (Cold spring harbor Laboratory Press, 1999); The Antibodies (M.Zaneti and J.D.Capra, eds., Harwood Academic Publishers, 1995); and Cancer: Principles and Practice of Oncology (V.T.DeVita et al, eds., J.B.Lippincott Company, 1993).
Definition of
An "antibody" is an immunoglobulin molecule capable of specifically binding a target, e.g., a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site located in the variable region of the immunoglobulin molecule. As used herein, the term includes not only intact polyclonal or monoclonal antibodies, but also fragments thereof (e.g., Fab ', F (ab')2Fv, dAb), single chain antibody (ScFv), mutants thereof, chimeric antibodies, diabodies (diabodies), fusion proteins comprising an antibody portion, and any other modified configuration of an immunoglobulin molecule comprising an antigen recognition site. Antibodies include any type of antibody, such as IgG, IgA, or IgM (or subclasses thereof), and the antibody need not be of any particular type. Depending on the antibody amino acid sequence of the constant region of its heavy chain, immunoglobulins can be assigned to different types. There are 5 major types of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these classes can be further subdivided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA 2. Corresponding to different typesThe heavy chain constant domains of the immunoglobulins of (a) are referred to as α, δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different types of immunoglobulins are well known.
"Fv" are antibody fragments that contain an intact antigen recognition and binding site. In the two-chain Fv class, this region consists of a dimer of one heavy and one light chain variable domain in tight, non-covalent association. In the single chain Fv class, a heavy chain variable domain and a light chain variable domain can be covalently linked by a flexible peptide linker such that the light and heavy chains can associate in a dimeric structure similar to that in the two chain Fv class. It is through this configuration that the 3 CDRs of each variable domain interact to determine the antigen binding specificity on the surface of the VH-VL dimer. However, even a single variable domain (or half of an Fv comprising only 3 CDRs specific for an antigen) has the ability to recognize and bind antigen, although usually with lower affinity than the entire binding site.
The Fab fragment also contains the constant domain of the light chain and the first constant domain of the heavy chain (CH 1). Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain, including one or more cysteines from the antibody hinge region. The f (ab)2 fragment is a bivalent fragment comprising 2 Fab fragments linked by disulfide bonds of the hinge region.
An antibody may have one or more binding sites (to bind to an antigen). If more than one binding site is present, the binding sites may be the same as each other or may be different. For example, a naturally occurring immunoglobulin has two identical binding sites, a single chain antibody or Fab fragment has one binding site, while a "bispecific" or "bifunctional" antibody (diabody) has two different binding sites in terms of sequence and/or antigen/epitope recognition.
An "isolated antibody" is (1) an antibody that does not bind to its accompanying natural binding components in its native state, including other naturally-bound antibodies, (2) an antibody that does not contain other proteins from the same species, (3) an antibody expressed by cells from a different species, or (4) a non-naturally-occurring antibody.
"monoclonal antibody" refers to a homogeneous population of antibodies, wherein the monoclonal antibody consists of amino acids (naturally occurring and non-naturally occurring) that are involved in the selective binding of an antigen. The population of monoclonal antibodies is highly specific, being directed against a single antigenic site. The term "monoclonal antibody" includes not only intact monoclonal antibodies and full-length monoclonal antibodies, but also fragments thereof (e.g., Fab ', F (ab')2Fv), single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of an immunoglobulin molecule comprising an antigen recognition site of the desired specificity and ability to bind antigen. No limitations are intended with respect to the source of the antibody or the manner in which it is produced (e.g., by hybridoma, phage selection, recombinant expression, transgenic animal, etc.).
As used herein, a "humanized" antibody refers to a form of non-human (e.g., murine) antibody that is a specific chimeric immunoglobulin, immunoglobulin chain, or fragment thereof (e.g., Fv, Fab ', F (ab')2Or other antigen binding subsequences of antibodies). For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a Complementarity Determining Region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and biological activity. In some cases, Fv Framework Region (FR) residues of the human immunoglobulin are replaced with corresponding non-human residues. In addition, humanized antibodies may comprise residues that are not found in the recipient antibody and the introduced CDR or framework sequences, but which are included to further improve and optimize antibody performance. In general, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regionsIs the FR region of the human immunoglobulin consensus sequence. The humanized antibody will also optimally include at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. The antibody may have a modified Fc region as described in WO 99/58572. Other forms of humanized antibodies have one or more CDRs (1, 2, 3,4, 5, 6) that have been altered relative to the original antibody, also referred to as one or more CDRs "derived" from one or more CDRs of the original antibody.
As used herein, "human antibody" refers to an antibody having an amino acid sequence corresponding to the amino acid sequence of an antibody produced by a human, and/or has been prepared using any of the techniques known in the art or disclosed herein for preparing human antibodies. This definition of human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide. One such example is an antibody comprising murine light chain and human heavy chain polypeptides. Human antibodies can be produced using various techniques known in the art. In one embodiment, the human antibody is selected from a phage library, wherein the phage library expresses human antibodies (Vaughan et al, 1996, Nature Biotechnology, 14: 309-. Human antibodies can also be prepared by introducing human immunoglobulin loci into transgenic animals, such as mice, in which endogenous immunoglobulin genes have been partially or fully inactivated. The process is described in us patent 5,545,807; 5,545,806; 5,569,825; 5,625,126, respectively; 5,633,425 and 5,661,016. Alternatively, human antibodies can also be prepared by immortalizing human B lymphocytes that produce antibodies against the target antigen (such B lymphocytes can be recovered from the individual or can have been immunized in vitro). See, e.g., Cole et al, Monoclonal antibodies and Cancer Therapy, Alan R.Liss, p.77 (1985); boerner et al, 1991, j.immunol., 147 (1): 86-95; and U.S. Pat. No. 5,750,373.
Single chain antibodies (scFc) are antibodies in which the VL and VH regions are paired via a synthetic linker that enables them to be prepared as a single protein chain to form monovalent molecules (Bird et al Science, 242: 423-.
A diabody is a bivalent, bispecific antibody in which the VH and VL domains are expressed on a single polypeptide chain and a linker is used which is too short to allow pairing between the 2 domains on the same chain, forcing the domains to pair with the complementary domains of the other chain and generating two antigen-binding sites.
"chimeric antibody" refers to an antibody in which a portion of each of the amino acid sequences of the heavy and light chains is homologous to the corresponding sequence in an antibody derived from a particular species or belonging to a particular class, while the remaining segments of the chains are homologous to the corresponding sequences in another antibody. Typically, in such chimeric antibodies, the variable regions of the light and heavy chains mimic those of an antibody derived from one mammalian species, while the constant portions are homologous to sequences in an antibody derived from another species. One obvious advantage of such chimeric forms is that, for example, the variable regions can be readily produced from currently known sources by using readily available hybridomas or B cells (which are derived from non-human host organisms), and the constant regions are derived from, for example, human cell preparations. While the variable region has the advantages of ease of preparation and specificity unaffected by its source, human-derived constant regions are less likely to elicit an immune response in a human subject when injected with an antibody than constant regions from non-human sources. However, the definition is not limited to the specific example.
A "functional Fc region" has at least one effector function of a native sequence Fc region. Exemplary "effector functions" include C1q binding; complement Dependent Cytotoxicity (CDC); fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis (phagocytosis); downregulation of cell surface receptors (e.g., B cell receptors; BCR), and the like. Such effector functions typically require an Fc region in combination with a binding domain (e.g., an antibody variable domain), and can be assessed using various assays known in the art for assessing such antibody effector functions.
"native sequence Fc region" includes amino acid sequences identical to the amino acid sequence of a naturally found Fc region. "variant Fc region" includes an amino acid sequence that differs from the amino acid sequence of a native sequence Fc region by at least one amino acid modification, but which still retains at least one effector function of the native sequence Fc region. Preferably, the variant Fc region has at least one amino acid substitution, such as from about 1 to about 10 amino acid substitutions, preferably from about 1 to about 5 amino acid substitutions, in the native sequence Fc region or in the Fc region of the parent polypeptide as compared to the native sequence Fc region or to the Fc region of the parent polypeptide. In this context, the variant Fc region preferably has at least about 80% sequence identity with the native sequence Fc region and/or with the Fc region of the parent polypeptide, more preferably at least about 90% sequence identity therewith, and more preferably at least about 95% sequence identity therewith.
As used herein, "antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (fcrs) (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on target cells, subsequently causing lysis of the target cells. ADCC activity of a molecule of interest can be assessed using an in vitro ADCC assay, such as the assay described in U.S. patent 5,500,362 or 5,821,337. Effector cells useful in such assays include Peripheral Blood Mononuclear Cells (PBMC) and NK cells. Alternatively or additionally, the ADCC activity of the molecule of interest may be measured, for example, in animal models such as Clynes et al, 1998, pnas (usa), 95: in vivo evaluation was performed in the animal model disclosed in 652-656.
As used herein, "Fc receptor" and "FcR" describe receptors that bind the Fc region of an antibody. A preferred FcR is a native sequence human FcR. In addition, a preferred FcR is one that binds an IgG antibody (gamma receptor) and includes receptors of the Fc γ RI, Fc γ RII, and Fc γ RIII subclasses, including allelic variants and alternatively spliced forms of such receptors. Fc γ RII receptors include Fc γ RIIA ("activating receptor") and Fc γ RIIB ("inhibiting receptor"), which have similar amino acid sequences (differing primarily in their cytoplasmic domains). FcR is reviewed in ravatch and Kinet, 1991, ann.rev.immunol., 9: 457-92; capel et al, 1994, immunoassays, 4: 25-34; and de Haas et al, 1995, j.lab.clin.med., 126: 330-41. "FcR" also includes the neonatal receptor FcRn, which is responsible for the transfer of maternal IgG to the fetus (Guyer et al, 1976, J.Immunol., 117: 587; and Kim et al, 1994, J.Immunol., 24: 249).
"complement-dependent cytotoxicity" and "CDC" refer to the lysis of a target in the presence of complement. The complement activation pathway is initiated by the binding of the first component of the complement system (C1q) to a molecule (e.g., an antibody) that is complexed with a related antigen. To assess complement activation, CDC assays can be performed, such as Gazzano-Santoro et al, j.immunol.methods, 202: 163 (1996).
As used herein, the terms "G1" and "antibody G1" are used interchangeably to refer to the antibody produced by the expression vectors with accession nos. ATCC-PTA-6867 and ATCC-PTA-6866. The amino acid sequences of the heavy and light chain variable regions are shown in SEQ ID Nos. 1 and 2. The CDR portions of antibody G1 (including Chothia and Kabat CDRs) are depicted in schematic form in fig. 5 of WO2007/054809 (the contents of which are incorporated herein by reference in their entirety). Polynucleotides encoding the heavy and light chain variable regions are shown in SEQ ID Nos. 9 and 10. The characteristics of antibody G1 are described in the examples of WO2007/054809, the entire contents of which are incorporated herein by reference. G1 is a humanized monoclonal blocking antibody (IgG2) that blocks the binding and activity of the neuropeptide CGRP (a and b) and its neurogenic vasodilatory effect caused by CGRP release. G1 is an IgG2 Δ a monoclonal anti-CGRP antagonist antibody derived from a murine anti-CGRP antagonist antibody precursor (designated muMAb7E9, as identified in a screen using splenocytes prepared from mice immunized with human and rat CGRP, fused with murine plasmacytoma cells). G1 was generated by grafting the CDRs of the muMAb7E 9-derived light and heavy chains into the nearest human germline sequence, followed by introduction of at least 1 mutation into each CDR and 2 framework mutations into the VH. Two mutations were introduced into the Fc domain of G1 to inhibit human Fc receptor activation. G1 and muMab7E9 have been shown to recognize the same epitope.
As used herein, the terms "G2" and "antibody G2" are used interchangeably and refer to an anti-rat CGRP mouse monoclonal antibody, such as Wong HC et al, human Hybridoma 12: 93-106 (1993). The amino acid sequences of the heavy and light chain variable regions are shown in SEQ ID Nos. 19 and 20. Polynucleotides encoding the heavy and light chain variable regions are shown in SEQ ID Nos. 27 and 28. The CDR portions of antibody G2 are provided in SEQ ID nos. 21 to 26. G2 has been shown to recognize the same epitope as G1.
As used herein, "immunospecific" binding of an antibody refers to an antigen-specific binding interaction that occurs between the antigen-binding site of an antibody and a particular antigen recognized by the antibody (i.e., the antibody reacts with a protein in an ELISA or other immunoassay, and does not react detectably with unrelated proteins).
"specifically binding" or "preferentially binding" (used interchangeably herein) to an epitope of an antibody or polypeptide is a term well known in the art, and methods for determining such specific or preferential binding are also well known in the art. A molecule is said to exhibit "specific binding" or "preferential binding" if it reacts or binds more frequently, more rapidly, with a longer duration, and/or with greater affinity to a particular cell or substance (as compared to its reaction or binding to another cell or substance). An antibody "specifically binds" or "preferentially binds" to a target if it binds the target with greater affinity, avidity, more readily, and/or for a longer duration of time (as compared to its binding to other substances). By reading this definition, it is also understood that, for example, an antibody (or portion or epitope) that specifically or preferentially binds a first target may or may not specifically or preferentially bind a second target. Thus, "specific binding" or "preferential binding" does not necessarily require (although it may include) specific binding. Typically, but not necessarily, binding refers to preferential binding.
The terms "polypeptide", "oligopeptide", "peptide" and "protein" are used interchangeably herein and refer to a polymer of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The term also includes amino acid polymers that have been modified either naturally or by intervention (intervention); for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation or any other manipulation or modification, such as conjugation to a labeling element. Also included in this definition are, for example, polypeptides that comprise one or more amino acid analogs (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It will be appreciated that, because the polypeptides of the invention are based on antibodies, the polypeptides may exist in single chain or conjugated chain form.
"polynucleotide" or "nucleic acid," as used interchangeably herein, refers to a polymer of nucleotides of any length, and includes DNA and RNA. The nucleotides may be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into the polymer by a DNA or RNA polymerase. Polynucleotides may comprise modified nucleotides such as methylated nucleotides and their analogs. Modifications to the nucleotide structure, if present, may be imparted before or after assembly of the polymer. The nucleotide sequence may be interrupted by non-nucleotide components. After polymerization, the polynucleotide may be further modified, for example by conjugation with a labeling element. Other types of modifications include, for example, "capping", replacement of one or more naturally occurring nucleotides with an analog, internucleotide modifications such as those using uncharged linkages (e.g., methylphosphonate, phosphotriester, phosphoamidate, carbamate, etc.) and those using charged linkages (e.g., phosphorothioate, phosphorodithioate, etc.), modifications containing pendant moieties such as proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), such thatModifications with intercalating agents (e.g., acridine, psoralen, and the like), modifications comprising chelators (e.g., metals, radioactive metals, boron, metal oxides, and the like), modifications comprising alkylating agents (alkylators), modifications having modified linkages (e.g., alpha anomeric (anomeric) nucleic acids, and the like), and unmodified forms of polynucleotides. Furthermore, any hydroxyl groups typically present in sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to create additional linkages to additional nucleotides, or may be conjugated to a solid support. The 5 and 3' terminal OH groups may be phosphorylated or may be replaced with organic capping group moieties of 1 to 20 carbon atoms or amines. Other hydroxyl groups may also be derivatized to standard protecting groups. Polynucleotides may also comprise analogous forms of ribose or deoxyribose known in the art, including, for example, 2 '-O-methyl-, 2' -O-allyl, 2 '-fluoro-or 2' -azido-ribose, carbocyclic sugar analogs, alpha-anomeric sugars, epimeric (epimeric) sugars such as arabinose, xylose or lyxose, pyranose, furanose, sedoheptulose, acyclic analogs, and alkali-free nucleoside analogs such as methylnucleosides. One or more phosphodiester linkages may be replaced with alternative linking groups. Such alternative linking groups include, but are not limited to, those wherein the phosphate ester is substituted with P (O) S ("thio)"), P (S) S ("dithio"), "(O) NR2("amidation"), P (O) R, P (O) OR', CO OR CH2("formacetal") where each R or R' is independently H or substituted or unsubstituted alkyl (1-20C) (optionally containing an ether (-O-) linkage), aryl, alkenyl, cycloalkyl, cycloalkenyl, or araldyl. Not all linkages in a polynucleotide need be identical. The above description applies to all polynucleotides mentioned herein, including RNA and DNA.
The "variable region" of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, alone or in combination. The variable regions of the heavy and light chains each consist of 4 Framework Regions (FRs) connected by 3 Complementarity Determining Regions (CDRs), also known as hypervariable regions. The CDRs in each chain are tightly bound together by the FRs and, together with the CDRs from the other chain, contribute to the formation of the antigen-binding site of the antibody. There are at least two techniques for determining CDRs: (1) methods based on sequence variability across species (i.e., Kabat et al Sequences of Proteins of immunological interest, (5 th edition, 1991, National Institutes of Health, Bethesda MD)); and (2) methods based on crystallographic studies of antigen-antibody complexes (Chothia et Al (1989) Nature 342: 877; Al-lazikani et Al (1997) J.Molec.biol.273: 927-948)). As used herein, a CDR may refer to a CDR determined by either method or by a combination of both methods.
The "constant region" of an antibody refers to either the antibody light chain constant region or the antibody heavy chain constant region, alone or in combination.
As used herein, an "anti-CGRP antagonist antibody" (interchangeably referred to as "anti-CGRP antibody") refers to an antibody that is capable of binding to CGRP and inhibiting CGRP biological activity and/or downstream pathways. anti-CGRP antagonist antibodies include antibodies that block, antagonize, inhibit, or reduce (including significantly) the biological activity of CGRP. For the purposes of the present invention, it is to be expressly understood that the term "anti-CGRP antagonist antibody" includes all the previously identified terms, names and functional states and characteristics whereby the CGRP itself, the biological activity of the CGRP or the consequences of the biological activity are substantially eliminated, reduced or neutralized to any meaningful degree. Examples of anti-CGRP antagonist antibodies are provided herein.
As used herein, "substantially pure" refers to a material that is at least 50% pure (i.e., free of contaminants), more preferably at least 90% pure, more preferably at least 95% pure, more preferably at least 98% pure, more preferably at least 99% pure.
"host cell" includes individual cells or cell cultures which may be or have been the recipient of the vector for integration of the polynucleotide insert. Host cells include progeny of a single host cell, and the progeny may not necessarily be identical (morphologically or on genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. Host cells include cells transfected in vivo with a polynucleotide of the invention.
As used herein, "treatment" is a method for obtaining a beneficial or desired clinical result. For purposes of the present invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: amelioration or reduction of inflammatory pain and/or any aspect of the symptoms of inflammatory pain. For purposes of the present invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: including relief, lessening severity of pain and/or symptoms associated with inflammatory pain.
An "effective amount" of a drug, compound or pharmaceutical composition is an amount sufficient to produce a beneficial or desired result, including a reduction or decrease in clinical outcome, such as pain sensation. An effective amount may be administered in one or more administrations. For the purposes of the present invention, an effective amount of a drug, compound or pharmaceutical composition is an amount sufficient to treat, ameliorate, reduce the intensity of and/or prevent inflammatory pain or a symptom associated with inflammatory pain. As understood in the clinical setting, an effective amount of a drug, compound or pharmaceutical composition may or may not be obtained in conjunction with another drug, compound or pharmaceutical composition. Thus, an "effective amount" may be considered in the context of administering one or more therapeutic agents, and an individual agent may be considered to be provided in an effective amount if the desired result is obtained or achievable in combination with one or more other agents.
In one embodiment, "prepared for" means herein that the agent is present in dosage units or the like, suitably packaged and/or labeled for peripheral administration.
By "decreasing incidence" of inflammatory pain and/or symptoms associated with inflammatory pain is meant any decreasing severity (which may include a decreasing need for and/or amount of other drugs and/or treatments typically used for such conditions), duration, and/or frequency.
By "ameliorating" inflammatory pain and/or symptoms associated with inflammatory pain is meant a reduction or amelioration of one or more symptoms of inflammatory pain and/or symptoms associated with inflammatory pain when compared to the absence of administration of an anti-CGRP antagonist antibody. "improvement" also includes a reduction or decrease in the duration of symptoms.
By "alleviating" inflammatory pain and/or symptoms associated with inflammatory pain is meant reducing the extent of one or more undesirable clinical manifestations of inflammatory pain in an individual or population of individuals treated with an anti-CGRP antagonist antibody according to the invention.
As used herein, "delaying" the progression of inflammatory pain means delaying, arresting, slowing, delaying, stabilizing and/or delaying the progression of inflammatory pain and/or symptoms associated with inflammatory pain. The delay may be of different lengths of time depending on the history of the disease and/or the individual to be treated. As will be apparent to those skilled in the art, a sufficient or significant delay may in fact include prevention, as the individual does not develop inflammatory pain. A method of "delaying" the development of a symptom is a method that reduces the likelihood of developing a symptom within a given time frame and/or reduces the extent of a symptom within a given time frame when compared to not using the method. Such comparisons are typically based on clinical studies (using statistically significant numbers of subjects).
"biological samples" include various sample types obtained from an individual and can be used in diagnostic or monitoring assays. This definition includes blood and other liquid samples of biological origin, solid tissue samples such as biopsy specimens or tissue cultures or cells derived therefrom, and the progeny thereof. The definition also includes samples that have been manipulated in any way after they have been obtained (e.g., by treatment with reagents, solubilization, or enrichment for certain components such as proteins or polynucleotides, or for sectioning, embedded in a semi-solid or solid matrix). The term "biological sample" includes clinical samples and also includes cells in culture, cell supernatants, cell lysates, serum, plasma, biological fluids, and tissue samples.
An "individual" or "subject" is a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, farm animals (e.g., cattle), sport animals (sport animals), pets (e.g., cats, dogs, and horses), primates, mice, and rats.
As used herein, "vector" refers to a construct capable of delivering and preferably expressing one or more genes or sequences of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents (condensing agents), DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells such as producer cells (producer cells).
As used herein, "expression control sequence" refers to a nucleic acid sequence that directs the transcription of a nucleic acid. The expression control sequence may be a promoter, such as a constitutive or inducible promoter, or an enhancer. The expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
As used herein, a "pharmaceutically acceptable carrier" includes any material that, when combined with an active ingredient, allows the ingredient to retain biological activity and not react with the immune system of a subject. Examples include, but are not limited to, any standard pharmaceutical carrier such as phosphate buffered saline, water, emulsions such as oil/water emulsions, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal saline (0.9%). Compositions comprising such carriers are formulated by well-known conventional methods (see, e.g., Remington's Pharmaceutical Sciences, 18 th edition, A. Gennaro, ed., Mack Publishing Co., Easton, PA, 1990; and Remington, The Science and Practice of Pharmacy, 20 th edition, Mack Publishing, 2000).
As used herein, the term "peripherally administered" refers to the route by which substances, agents and/or anti-CGRP antagonist antibodies are delivered, particularly it refers to non-central, non-spinal, non-intrathecal, non-direct delivery into the CNS. The term refers to routes of administration other than those just described, including oral, sublingual, buccal, topical (topical), rectal, by inhalation, transdermal, subcutaneous, intravenous, intraarterial, intramuscular, intracardiac, intraosseous, intradermal, intraperitoneal, transmucosal, transvaginal, intravitreal, intraarticular, periarticular, local (local) or epicutaneous (epicutaneous) routes of administration.
The term "peripherally acting", as used herein, refers to the site of action of a substance, compound, agent and/or anti-CGRP antagonist antibody within the peripheral nervous system relative to the central nervous system, which compound, agent and/or anti-CGRP antagonist antibody is restricted from passing through the barrier to the CNS and brain when administered peripherally. The term "central penetration" refers to the ability of a substance to cross a barrier to reach the brain or CNS.
The term "Koff", as used herein, means the dissociation rate constant for dissociation of an antibody from an antibody/antigen complex.
The term "Kd", as used herein, means the dissociation constant of an antibody-antigen interaction.
The present invention relates to agents for preventing and/or treating inflammatory pain and/or symptoms of inflammatory pain in an individual and methods for preventing and/or treating inflammatory pain and/or symptoms of inflammatory pain in an individual.
In a first aspect, the invention provides the use of an anti-CGRP antagonist antibody for the manufacture of a medicament for the prevention and/or treatment of inflammatory pain and/or symptoms of inflammatory pain, wherein the medicament is prepared for peripheral administration or wherein the medicament is administered peripherally.
In a second aspect, the invention provides an anti-CGRP antagonist antibody for use in the prevention and/or treatment of inflammatory pain and/or symptoms of inflammatory pain, wherein the antibody is prepared for peripheral administration or wherein the antibody is administered peripherally.
In a third aspect, the present invention provides the use of an anti-CGRP antagonist antibody for the manufacture of a medicament for ameliorating, controlling, reducing the incidence of or delaying the development or progression of inflammatory pain and/or symptoms of inflammatory pain, wherein the medicament is prepared for peripheral administration or wherein the medicament is administered peripherally. In an alternative aspect of this aspect, the invention provides an anti-CGRP antagonist antibody for use in ameliorating, controlling, reducing the incidence of or delaying the development or progression of inflammatory pain and/or symptoms of inflammatory pain, wherein the antibody is prepared for peripheral administration or wherein the antibody is administered peripherally.
In a fourth aspect, the present invention provides a method of preventing and/or treating inflammatory pain and/or symptoms of inflammatory pain in an individual, the method comprising peripherally administering to the individual an effective amount of an anti-CGRP antagonist antibody.
In a fifth aspect, the invention provides a method of ameliorating, controlling, reducing the incidence of, or delaying the development or progression of inflammatory pain and/or symptoms of inflammatory pain in an individual, the method comprising peripherally administering to the individual an effective amount of an anti-CGRP antagonist antibody.
According to a preferred embodiment of the invention, the individual is preferably a mammal, for example a companion animal such as a horse, cat or dog or a livestock animal such as a sheep, cow or pig. Most preferably, the mammal is a human.
According to a preferred embodiment of the invention, the medicament and/or the anti-CGRP antagonist antibody is prepared for oral, sublingual, buccal, topical, rectal, inhalation, transdermal, subcutaneous, intravenous, intra-arterial, intramuscular, intracardiac, intraosseous, intradermal, intraperitoneal, transmucosal, vaginal, intravitreal, intra-articular, periarticular, local or epicutaneous administration.
According to other preferred embodiments, the medicament is prepared for peripheral administration before and/or during and/or after the development of inflammatory pain.
In one embodiment, the anti-CGRP antagonist antibody acts peripherally after administration. In one embodiment, the anti-CGRP antagonist antibody is not administered by the central, spinal, or intrathecal route.
According to a preferred embodiment of the invention, the inflammatory pain is arthritis pain, which may be rheumatoid arthritis pain or osteoarthritis pain. Preferably, the inflammatory pain is osteoarthritis pain.
The use and method of the present invention may be used to improve the physical function and/or stiffness of an individual suffering from osteoarthritis.
The diagnosis or assessment of rheumatoid arthritis pain is well established in the art. The assessment may be based on measurement methods known in the art, such as patient pain characterization using various pain scales (pain scales). See, e.g., Katz et al, Surg Clin North Am (1999)79 (2): 231-52; caraceni et al J Pain Symptom Manage (2002)23 (3): 239-55. There are also common scales for measuring disease status such as American College of Rheometric (ACR) (Felson et al, Arthritis and Rheometric (1993)36 (6): 729-.
In one embodiment, one or more of ACR, HAQ and AIMS are utilized to measure the improvement, control, reduced incidence or delay in development or progression of rheumatoid arthritis pain and/or symptoms of rheumatoid arthritis pain.
The diagnosis or assessment of osteoarthritis pain is also well established in the art. The assessment can be made based on measurement methods known in the art, such as patient pain characterization using various pain scales. See, e.g., Katz et al, Surg Clin North Am (1999)79 (2): 231-52; caraceni et al J Pain Symptom Manage (2002)23 (3): 239-55. For example, the WOMAC ambulatory Pain Scale (including Pain, stiffness, and physical function) and the 100mm Visual Analog Scale (VAS) can be used to assess Pain and estimate response to treatment.
In one embodiment, the improvement, control, reduced incidence, or delay in development or progression of osteoarthritis pain and/or symptoms of osteoarthritis pain is measured using one or more of a WOMAC ambulatory pain dose table and a VAS.
According to a preferred embodiment of the invention, the anti-CGRP antagonist antibody binds to CGRP, more preferably to CGRP and inhibits the ability of CGRP to bind to CGRP receptor. Preferably, the anti-CGRP antagonist antibody binds to human and rodent CGRP, preferably human and rat CGRP. More preferably the antibody binds to human CGRP, more preferably the anti-CGRP antagonist antibody binds to human α -CGRP or to human α -CGRP and/or β -CGRP. Most preferably, the anti-CGRP antagonist antibody is an antibody that exhibits any one or more of the following functional characteristics: (a) combining CGRP; (b) preventing CGRP from binding to its receptor; (c) block or reduce activation of CGRP receptors, including cAMP activation; (d) inhibiting, blocking, suppressing, or reducing the biological activity of CGRP, including downstream pathways mediated by CGRP signaling, such as receptor binding and/or induction of a cellular response to CGRP; (e) any aspect that prevents, ameliorates or treats inflammatory pain; (f) increasing CGRP clearance; and (g) inhibiting (reducing) the synthesis, production or release of CGRP.
Antibodies of the invention (including G1 and G2) are known to bind CGRP and abrogate its bioavailability, e.g., in serum, thereby preventing CGRP from contacting its receptor and the downstream cellular responses and biological effects of CGRP such as flushing and vasodilation.
In further preferred embodiments of the invention, the anti-CGRP antagonist antibody binds to a fragment of CGRP, more preferably to a fragment of CGRP as well as to full-length CGRP. Preferably, the anti-CGRP antagonist antibody binds to the C-terminal region or fragment of CRGP. The C-terminal region or fragment of CRGP preferably comprises amino acids 19-37 or 25-37 or 29-37 or alternatively 30-37, yet alternatively 31-37 of CGRP. In a further embodiment, the C-terminal region or fragment of CRGP preferably comprises amino acids 32-37, most preferably 33 to 37, of CGRP. Preferably the CGRP is an alpha-CGRP or a beta-CGRP, more preferably a human or a rodent, more preferably a human or a rat, more preferably a human alpha-CGRP or a beta-CGRP, most preferably a human alpha-CGRP.
In a further preferred embodiment of the invention, the anti-CGRP antagonist antibody specifically binds to the amino acid sequence GSKAF. Preferably, the sequence GSKAF of CGRP is the epitope bound by the anti-CGRP antagonist antibody, preferably at positions 33 to 37, most preferably the sequence is gxxf (where X can be any amino acid), preferably at positions 33 to 37 of CGRP, the ends of which are defined by amino acids G33 and F37 of CGRP.
In one embodiment, the invention provides anti-CGRP antagonist antibodies that specifically bind to an epitope defined by amino acids G33 through F37 of CGRP. The anti-CGRP antagonist antibody can specifically bind to an epitope defined by the amino acid sequence GSKAF. In one embodiment, the invention provides the use of such antibodies in the applications and methods defined in the various aspects of the invention.
In one embodiment, the anti-CGRP antagonist antibody inhibits or prevents activation of the CGRP receptor. Preferably, the anti-CGRP antibody has an IC of 0.0001(0.1nM) to 500. mu.M50. In some preferred embodiments, IC50Is any one of 0.0001 μ M to 250 μ M, 100 μ M, 50 μ M, 10 μ M, 1 μ M, 500nM, 250nM, 100nM, 50nM, 20nM, 15nM, 10nM, 5nM, 1nM, or 0.5nM, or is about any one of 250 μ M, 100 μ M, 50 μ M, 10 μ M, 1 μ M, 500nM, 250nM, 100nM, 50nM, 20nM, 15nM, 10nM, 5nM, 1nM, or 0.5nM, as measured in an in vitro binding assay. In some further preferred embodiments, the IC50Less than 500pM or 100pM or 50pM, as measured in an in vitro binding assay. In another more preferred embodiment, the IC50Is 1.2nM or 31 nM.
In a further preferred embodiment, the anti-CGRP antagonist antibody used is capable of competing with the antibodies described herein above for binding to CGRP or to a fragment of CGRP, or to a fragment of CGRP as well as to full length CGRP, preferably to the C-terminal region or fragment of CRGP, preferably the C-terminal region or fragment of CRGP comprises amino acids 19-37 or 25-37 or 29-37 or alternatively 30-37 or alternatively 31-37 of CGRP. In a further embodiment, the C-terminal region or fragment of CRGP preferably comprises amino acids 32-37, most preferably 33 to 37, of CGRP.
In a further preferred embodiment, the anti-CGRP antagonist antibody or antigen-binding portion thereof as used in the present invention is capable of binding to an anti-CGRP antagonist antibody as described herein above, in particular as described herein as G1 or G2:
(a) (ii) competes for binding to CGRP or a fragment of CGRP, or to fragments of CGRP as well as to full-length CGRP, preferably to the C-terminal region or fragment of CRGP, preferably the C-terminal region or fragment of CRGP comprises amino acids 19-37 or 25-37 or 29-37 or alternatively 30-37, additionally alternatively 31-37, preferably amino acids 32-37, most preferably 33 to 37, of CGRP, preferably CGRP is alpha or beta CGRP, preferably beta CGRP, more preferably rodent or human CGRP, most preferably human CGRP,
(b) competing for binding to the epitope sequence GSKAF, which is preferably at amino acid positions 33 to 37 of the CGRP defined in (a), more preferably to the sequence GXXFF, wherein X is any amino acid, preferably GXXFF is at amino acid positions 33 to 37 of the CGRP defined in (a),
(c) with substantially the same Kd and/or substantially the same KoffCompete for binding as described in (a) or (b), and/or
(d) Competes for binding to CGRP and inhibits/antagonizes CGRP biological activity and/or downstream pathways, preferably CGRP is alpha or beta CGRP, preferably beta CGRP, more preferably rodent or human CGRP, most preferably human CGRP.
The anti-CGRP antagonist antibody is preferably used at 0.000001. mu.M (0.001nM) or 0.00001. mu.M (0.01nM) to 50Binding affinity (K) of 0. mu.Md) Binding to CGRP, a region of CGRP or a fragment of CGRP. In some preferred embodiments, the binding affinity (Kd) is between 0.000001 μ M or 0.00001 μ M and any of 250 μ M, 100 μ M, 50 μ M, 10 μ M, 1 μ M, 500nM, 250nM, 100nM, 50nM, 20nM, 15nM, 10nM, 5nM, 1nM, 0.5nM, 1nM, 0.05nM or 0.01nM, 0.005nM, 0.001nM, or is about any of 250 μ M, 100 μ M, 50 μ M, 10 μ M, 1 μ M, 500nM, 250nM, 100nM, 50nM, 20nM, 15nM, 10nM, 5nM, 1nM, 0.5nM, 1nM, 0.05nM, or 0.01nM, 0.005nM, 0.001nM, as measured in an in vitro binding assay. In some further preferred embodiments, the binding affinity (Kd) is less than 500pM or any of 100pM, 50pM or 10pM, 5pM, 1pM, as measured in an in vitro binding assay. In yet further more preferred embodiments, the binding affinity (Kd) is 0.04nM or 16 nM.
The anti-CGRP antagonist antibody used in the present invention may be selected from: monoclonal antibodies, polyclonal antibodies, antibody fragments (e.g., Fab ', F (ab')2Fv, Fc, ScFv, etc.), chimeric antibodies, bispecific antibodies, heteroconjugate antibodies (heteroconjugate antibodies), single chain (ScFv) antibodies, mutants thereof, fusion proteins comprising an antibody portion (e.g., domain antibodies), humanized antibodies, and any other modified configuration of an immunoglobulin molecule comprising an antigen recognition site of a desired specificity, including glycosylated variants of an antibody, amino acid sequence variants of an antibody, and covalently modified antibodies. The anti-CGRP antagonist antibody may be murine, rat, human or any other source (including chimeric or humanized antibodies). In some embodiments, the anti-CGRP antagonist antibody may be humanized, but more preferably is a human antibody. Preferably, the anti-CGRP antagonist antibody is an isolated antibody, more preferably it is a substantially pure antibody. When the anti-CGRP antagonist antibody is an antibody fragment, the fragment preferably retains the functional characteristics of the original antibody, i.e., CGRP binding and/or antagonist activity as described in the functional characteristics above.
Examples of anti-CGRP antagonist antibodies are known in the art. Thus according to a preferred embodiment of the invention, the anti-CGRP antagonist antibody used in the present invention is preferably an anti-CGRP antibody as described in (i) WO2007/054809, (ii) WO2007/076336, (iii) Tan et al, clin.sci. (Lond) 89: 565-73, 1995, (iv) Sigma (Missouri, US), product number C7113 (clone #4901), (v) Plourde et al, Peptides 14: an anti-CGRP antibody as disclosed generally or specifically in any one of 1225-1229, 1993 or which comprises or consists of:
(a) fragments of said antibodies (e.g., Fab ', F (ab')2Fv, Fc, ScFv, etc.),
(b) the light chain of the antibody is selected from the group consisting of,
(c) the heavy chain of the antibody is selected from the group consisting of,
(d) one or more variable regions from the light chain and/or heavy chain of said antibody,
(e) one or more CDRs (1, 2, 3,4, 5 or 6 CDRs) of the antibody,
(f) CDR H3 from the heavy chain of said antibody,
(g) CDR L3 from the light chain of said antibody,
(h) 3 CDRs from the light chain of the antibody,
(i) 3 CDRs from the heavy chain of the antibody,
(j) 3 CDRs from the light chain of the antibody and 3 CDRs from the heavy chain of the antibody,
(k) any one or more of (a) to (j).
According to a preferred embodiment of the invention, the anti-CGRP antagonist antibody is antibody G2 or antibody G1. According to the most preferred embodiments of the present invention, the anti-CGRP antagonist antibody used is the anti-CGRP antibody G1 specifically disclosed in patent application WO2007/054809, or includes variants thereof shown in table 6 of WO2007/054809, also including functionally equivalent antibodies of G1, i.e. comprising conservative substitutions of amino acid residues or deletions or additions of one or more amino acids that do not significantly affect their functional characteristics (e.g. CRGP binding or antagonist activity), as well as variants with enhanced or reduced activity and/or binding. As used herein, the terms "G1" and "antibody G1" are used interchangeably and refer to the antibody produced by the expression vector disclosed in application WO2007/054809 with accession numbers ATCCPTA-6867 and ATCC PTA-6866.
According to a further embodiment of the invention, the anti-CGRP antagonist antibody comprises or consists of a polypeptide selected from the group consisting of: (a) antibody G1 or a variant thereof shown in table 6 of WO 2007/054809; (b) a fragment or region of antibody G1 or a variant thereof shown in table 6 of WO 2007/054809; (c) the light chain of antibody G1 or a variant thereof shown in table 6 of WO 2007/054809; (d) the heavy chain of antibody G1 or a variant thereof shown in table 6 of WO 2007/054809; (e) one or more variable regions of the light chain and/or heavy chain of antibody G1 or a variant thereof shown in table 6 from WO 2007/054809; (f) one or more CDRs (1, 2, 3,4, 5 or 6 CDRs) of antibody G1 or a variant thereof shown in table 6 of WO 2007/054809; (g) CDR H3 of the heavy chain of antibody G1 or a variant thereof shown in table 6 from WO 2007/054809; (h) CDR L3 of the light chain of antibody G1 or a variant thereof shown in table 6 from WO 2007/054809; (i) 3 CDRs from the light chain of antibody G1 or variants thereof shown in table 6 of WO 2007/054809; (j) 3 CDRs from the heavy chain of antibody G1 or variants thereof shown in table 6 of WO 2007/054809; (k) 3 CDRs of the light chain and/or 3 CDRs of the heavy chain from antibody G1 or variants thereof shown in table 6 of WO 2007/054809; and (i) an antibody comprising any one of (b) to (k). The invention also provides a polypeptide comprising any one or more of the above. In some embodiments, at least 1, 2, 3,4, 5, or 6 CDRs are at least about 85%, 86%, 87%, 88%, 89%, 90%, 95%, 96%, 97%, 98%, or 99% identical to at least 1, 2, 3,4, 5, or 6 CDRs of G1 or variants thereof shown in table 6 of WO 2007/054809.
Determination of CDR regions is well within the ability of those skilled in the art. It is understood that in some embodiments, the CDRs may be a combination of Kabat and Chothia CDRs. In some embodiments, the CDRs are Kabat CDRs. In other embodiments, the CDR is a Chothia CDR.
An anti-CGRP antagonist antibody preferably comprises a fragment or region of antibody G1 or a variant thereof shown in table 6 of WO2007/054809 (e.g., Fab ', F (ab')2Fv, Fc, ScFv, etc.) or consists of said fragment or region. Preferably, the fragment or region has the functional characteristics of an anti-CGRP antagonist antibody, e.g. CGRP binding activity and/or antagonist activity, and comprises or consists of one or more light chains, heavy chains, fragments comprising one or more variable regions from a light chain and/or heavy chain, or one or more CDRs from a light chain and/or heavy chain of antibody G1.
According to a further preferred embodiment of the invention, the anti-CGRP antagonist antibody comprises a light chain variable region LCVR and/or a heavy chain variable region HCVR, said light chain variable region comprising a light chain variable region LCVR having the amino acid sequence selected from SEQ ID nos: 28-32, said heavy chain variable region comprising a peptide having a sequence selected from SEQ ID nos: 34-38.
More preferably, the anti-CGRP antagonist antibody comprises an LCVR polypeptide of SEQ ID NO shown in table 1 of patent application WO2007/076336 and further comprises an HCVR polypeptide of SEQ ID NO shown in table 1 of patent application WO 2007/076336.
According to a further embodiment of the invention, the anti-CGRP antagonist antibody used comprises an amino acid sequence selected from SEQ ID nos: 8-13 and/or a light chain cdr (cdrl) selected from SEQ ID No: 14-22 heavy chain CDR (CDRH).
Methods for the preparation and isolation of anti-CGRP antagonist antibodies of application WO2007/076336 and data demonstrating CGRP binding and antagonist characteristics of said antibodies are described in application WO 2007/076336.
Preferably, the anti-CGRP antagonist antibody for use in the present invention comprises a VH domain which is identical to SEQ ID NO: 1 or SEQ ID NO: 19 is at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical in amino acid sequence.
Preferably, the anti-CGRP antagonist antibody comprises a VL domain that is identical to SEQ ID NO: 2 or SEQ ID NO: 20 is at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical in amino acid sequence.
The anti-CGRP antagonist antibody preferably comprises a VH domain and a VL domain that are amino acid sequence identical to SEQ ID NO: 1 and 2 or a sequence corresponding to SEQ ID NO: 19 and 20 are at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.
Preferably, the anti-CGRP antagonist antibody comprises an amino acid sequence that is identical to seq id NO: 1 and a VH domain at least 90% identical in amino acid sequence to SEQ ID NO: 2 a VL domain that is at least 90% identical.
Alternatively, the anti-CGRP antagonist antibody preferably comprises an amino acid sequence that is identical to SEQ ID NO: 19 and a VH domain at least 90% identical in amino acid sequence to SEQ ID NO: 20 a VL domain that is at least 90% identical.
The anti-CGRP antagonist antibody preferably comprises at least one CDR selected from: (a) SEQ ID NO: 3 or 21, CDR H1; (b) the amino acid sequence of SEQ ID NO: CDRH2 as shown in 4 or 22; (c) the amino acid sequence of SEQ ID NO: CDR H3 shown in 5 or 23; (d) the amino acid sequence of SEQ ID NO: CDR L1 shown in FIG. 6 or 24; (e) SEQ ID NO: CDR L2 shown in 7 or 25; (f) SEQ ID NO: CDR L3 shown in 8 or 26; variants of CDR L1, CDR L2, and CDR H2 shown in table 6 of WO 2007/054809.
According to a preferred embodiment of the invention, the anti-CGRP antagonist antibody heavy chain constant region may be from any type of constant region, such as IgG, IgM, IgD, IgA, and IgE; and any isotype such as IgG1, lgG2, IgG3, and IgG 4.
More preferably, the anti-CGRP antagonist antibody comprises a heavy chain produced by the expression vector with ATCC accession No. PTA-6867. More preferably, the anti-CGRP antagonist antibody comprises a light chain produced by the expression vector having ATCC accession No. PTA-6866. More preferably, the anti-CGRP antagonist antibody is produced by an expression vector having ATCC accession nos. PTA-6867 and PTA-6866.
Preferably, the anti-CGRP antagonist antibody for use in the present invention is antibody G1 or antibody G2 as defined herein.
According to further embodiments of the invention, the anti-CGRP antagonist antibody comprises a modified constant region, as described, for example, in WO 2007/054809. Preferably, the modified constant region is immunologically inert (including being partially immunologically inert) so that it does not cause complement-mediated lysis, does not stimulate antibody-dependent cell-mediated cytotoxicity (ADCC), and does not activate microglia. Preferably one or more of such activities of the modified constant region is reduced. Most preferably, the constant region is as defined in eur.j.immunol. (1999) 29: 2613-2624; PCT application No. PCT/GB 99/01441; and/or as described in uk patent application 9809951.8. According to a preferred embodiment of the invention, the anti-CGRP antagonist antibody comprises the human heavy chain IgG2 constant region comprising the following mutations: a330, P331 to S330, S331 (amino acid numbering refers to wild type IgG2 sequence). Eur.j.immunol. (1999) 29: 2613-2624.
Methods for the preparation and isolation of anti-CGRP antagonist antibodies of application WO2007/054809 and data demonstrating CGRP binding and antagonist characteristics of said antibodies are described in application WO 2007/054809. The sequences of SEQ ID nos. 1 to 14 of said application are provided herein as SEQ ID nos. 1 to 14, respectively.
According to a further embodiment of the invention, the medicament is prepared for peripheral administration 1 to 7 times per week, more preferably 1 to 4 times per month, more preferably 1 to 6 times per 6-month period, more preferably 1 to 12 times per year. Preferably, the medicament is prepared for peripheral administration over a period selected from: 1 time per day, 1 time per 2, 3,4, 5 or 6 days, 1 time per week, 1 time per 2 weeks, 1 time per 3 weeks, 1 time per month, 1 time per 2 months, 1 time per 3 months, 1 time per 4 months, 1 time per 5 months, 1 time per 6 months, 1 time per 7 months, 1 time per 8 months, 1 time per 9 months, 1 time per 10 months, 1 time per 11 months or 1 time per year. According to a preferred embodiment, the medicament is prepared for peripheral administration by a route selected from one or more of the following: oral, sublingual, buccal, topical, rectal, by inhalation, transdermal, subcutaneous, intravenous, intraarterial, or intramuscular, by intracardiac administration, intraosseous, intradermal, intraperitoneal, transmucosal, vaginal, intravitreal, transdermal, intraarticular, periarticular, or local.
According to a further embodiment of the invention, the medicament is prepared for peripheral administration at an antibody concentration of 0.1 to 200 mg/ml; preferably, the antibody concentration is any of about 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200mg/ml +/-10% error or between 0.1 and any of about 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200mg/ml +/-10% error, most preferably 50 mg/ml.
According to a further embodiment of the invention, the medicament is prepared for peripheral administration at an antibody concentration of 0.1 to 200mg/kg body weight; preferably, the antibody concentration is any one of approximately 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200mg/kg body weight +/-10% error or between 0.1 and any one of approximately 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200mg/kg body weight +/-10% error, most preferably at an antibody concentration of 10 mg/kg.
According to a preferred embodiment of the invention, the anti-CGRP antagonist antibody has more than 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 206, 210, more preferably more than 1, 8, more than 1, more preferably more than +/-7, more than 1, more than 8 days, 9. An in vivo half-life of any one of 10, 11 or 12 months.
Preferably, the anti-CGRP antagonist antibody has an in vivo half-life of more than 6 days.
According to a further preferred embodiment of the invention, the agent and/or the anti-CGRP antagonist antibody does not produce effects of central nervous system and/or cognitive impairment. Preferably, the agent and/or anti-CGRP antagonist antibody does not induce any one or more of the following: amnesia, confusion, personality disorganization, hypoesthesia (hypesthesia), abnormal thinking, trismus, vertigo, akathisia, apathy, ataxia, paraesthesia, CNS irritation, emotional lability, euphoria, hallucinations, hostility, hyperesthesia, hyperkinesia, hypotonia, dyskinesia, libido incrasse, mania, myoclonus, neuralgia, neuropathy, psychosis, seizure, abnormal talking, stupor, and suicidal ideation; dizziness, somnolence, insomnia, anxiety, tremor, depression or paresthesia. Most preferably, the agent and/or anti-CGRP antagonist antibody does not induce impairment of motor coordination or attention.
According to further embodiments of the invention, the agent and/or anti-CGRP antagonist antibody does not produce respiratory, renal or gastrointestinal damage.
According to further embodiments of the invention, the agent and/or anti-CGRP antagonist antibody does not produce a physically and/or psychologically dependent effect. Preferably, the agent and/or anti-CGRP antagonist antibody does not exhibit opiate, benzodiazepinePhencyclidine (PCP) or N-methyl-D-aspartate (NMDA) receptors or CNS stimulants, or produce any sedative or euphoric effect.
In one embodiment, the anti-CGRP antagonist antibody ameliorates, controls, reduces the incidence of, or delays the development or progression of central pain sensation after administration.
In another embodiment, the effect of the anti-CGRP antagonist antibody is equal to and/or superior to that of NSAIDS and/or opiates in the same model of inflammatory pain. In one embodiment, the anti-CGRP antagonist antibody is effective in treating a refractory pain population.
According to a further aspect of the invention there is provided a use or method according to any other aspect of the invention wherein the anti-CGRP antagonist antibody is combined with one or more additional pharmaceutically active compounds or agents, preferably for the treatment of inflammatory pain, and administered separately, sequentially or simultaneously. Preferably, the additional agent is selected from one or more of the following:
(i) opioid analgesics such as morphine, heroin, hydromorphone, oxymorphone, levorphanol, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, dextropropoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine, or pentazocine;
(ii) non-steroidal anti-inflammatory drugs (NSAIDs) such as aspirin, diclofenac, diflunisal (diflusinal), etodolac, fenbufen, fenoprofen, flufenil, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamic acid, mefenamic acid, nabumetone, naproxen, oxaprozin, phenylbutazone, piroxicam, sulindac, tolmetin or zomepirac, cyclooxygenase-2 (COX-2) inhibitors, celecoxib; rofecoxib; meloxicam; JTE-522; l-745, 337; NS 398; or a pharmaceutically acceptable salt thereof;
(iii) barbiturates, e.g. amobarbital, alprenol, sec-barbital, butabarbital, mebendal, methobarbital, pentobarbital, phenobarbital, secobarbital, talbarbital, thiopentobarbital or thiobarbital or a pharmaceutically acceptable salt thereof
(iv) Benzodiazepines with sedative effectSuch as chlordiazepoxide, chlordiazepoxideDiazepam, flurazepam, lorazepam, oxazepam, temazepam or triazolam or a pharmaceutically acceptable salt thereof;
(v) h1 antagonists with sedative effects, such as diphenhydramine, mepyramine, promethazine, chlorphenamine or clocycline or pharmaceutically acceptable salts thereof;
(vi) sedatives such as glutethimide, meprobamate, methaqualone or chloralbyrin or pharmaceutically acceptable salts thereof;
(vii) skeletal muscle relaxants, such as baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine, methocarbamol, or oxyphennaredine (orphrensine), or pharmaceutically acceptable salts thereof;
(viii) NMDA receptor antagonists, such as dextromethorphan ((+) -3-hydroxy-N-methyl morphinan) or its metabolite dextrorphan ((+) -3-hydroxy-N-methyl morphinan), ketamine, memantine, pyrroloquinoline quinone, or cis-4- (phosphonomethyl) -2-piperidinecarboxylic acid, or a pharmaceutically acceptable salt thereof;
(ix) α -adrenergic agents, such as doxazosin, tamsulosin, clonidine or 4-amino-6, 7-dimethoxy-2- (5-methanesulfonamido-1, 2, 3, 4-tetrahydroisoquinol-2-yl) -5- (2-pyridyl) quinazoline;
(x) Tricyclic antidepressants, such as desipramine, imipramine, amitriptyline (amytriptiline) or nortriptyline (nortriptiline);
(xi) Anticonvulsants, such as carbamazepine or valproate;
(xii) Tachykinin (NK) antagonists, in particular NK-3, NK-2 or NK-1 antagonists, such as (alpha R, 9R) -7- [3, 5-bis (trifluoromethyl) benzyl ] -8, 9, 10, 11-tetrahydro-9-methyl-5- (4-tolyl) -7H- [1, 4] diazocino [2, 1-g ] [1, 7] naphthyridine-6-13-dione (TAK-637), 5- [ [ (2R, 3S) -2- [ (1R) -1- [3, 5-bis (trifluoromethyl) phenyl ] ethoxy-3- (4-fluorophenyl) -4-morpholinyl ] methyl ] -1, 2-dihydro-3H-1, 2, 4-triazol-3-one (MK-869), lanopiptan, dapitaptan or 3- [ [ 2-methoxy-5- (trifluoromethoxy) phenyl ] methylamino ] -2-phenyl-piperidine (2S, 3S);
(xiii) Muscarinic antagonists such as oxybutynin (oxybutin), tolterodine, propiverine, trospium chloride (tropsium chloride), or darifenacin;
(xiv) COX-2 inhibitors, such as celecoxib, rofecoxib, or valdecoxib;
(xv) Non-selective COX inhibitors (preferably with GI protection), such as nitroflurbiprofen (HCT-1026);
(xvi) Coal tar analgesics (coal tar analgesic), in particular acetaminophen;
(xvii) Neuroleptic agents such as droperidol;
(xviii) A vanilloid receptor agonist (e.g., resiniferatoxin) or antagonist (e.g., capsaicinoid);
(xix) Beta-adrenergic agents such as propranolol;
(xx) Local anesthetics, such as mexiletine;
(xxi) Corticosteroids, such as dexamethasone;
(xxii) A 5-hydroxytryptamine receptor agonist or antagonist;
(xxiii) A cholinergic (nicotinic) analgesic;
(xxiv) Tramadol (trade mark);
(xxv) PDEV inhibitors, such as sildenafil, vardenafil or tadalafil (taladafil);
(xxvi) Alpha-2-delta ligands such as gabapentin or pregabalin;
(xxvii) Cannabinoids (canabinoids); and
(xxviii) Antidepressants, such as amitriptyline (Elavil), trazodone (Desyrel) and imipramine (tofranel) or anticonvulsants such as phenytoin (Dilantin) or carbamazepine (Tegretol).
According to a further aspect of the present invention there is provided a pharmaceutical composition for preventing and/or treating inflammatory pain and/or symptoms of inflammatory pain or for ameliorating, controlling or reducing the incidence of or delaying the development or progression of inflammatory pain and/or symptoms of inflammatory pain in an individual, the pharmaceutical composition comprising an anti-CGRP antagonist antibody and a pharmaceutically acceptable carrier and/or excipient, wherein the composition is prepared for peripheral administration.
According to a further aspect of the present invention, there is provided a kit comprising:
(a) a pharmaceutical composition as defined above; and
(b) instructions for peripherally administering to the individual an effective amount of the pharmaceutical composition to prevent and/or treat inflammatory pain and/or symptoms of inflammatory pain or ameliorate, control, reduce the incidence of, or delay the development or progression of inflammatory pain and/or symptoms of inflammatory pain.
The kit may comprise one or more containers containing an anti-CGRP antagonist antibody or polypeptide as described herein and instructions for use according to any of the methods and uses of the invention. The kit may further comprise a description of selecting an individual suitable for treatment based on identifying whether the individual has or is at risk of having inflammatory pain. Instructions for peripheral administration of a pharmaceutical composition can include information about the dosage, dosing regimen, and route of administration for the desired treatment.
The preferred features of each aspect of the invention are equally applicable to each other aspect mutatis mutandis.
Examples
The invention will now be described with reference to the following examples which are intended to illustrate, but not to limit the invention.
The following examples and figures were generated with reference to antibody G1 (anti-human CGRP human monoclonal antibody) and antibody G2 (anti-mouse CGRP mouse monoclonal antibody) (Wong HC et al Hybridoma 12: 93-106 (1993)).
Example 1: osteoarthritis (OA) mechanical pain model (transection of Medial Meniscus (MMT))
Transection (MMT) of the medial meniscus in one of the posterior knees (hind knee) of rats resulted in the development of cartilage damage and other joint changes similar to those that occurred in OA. The resulting joint pain leads to a persistent change in weight bearing in the hind limb of the rat (assessed using an interpupillary balance pain tester) and the development of mechanical (tactile) allodynia (assessed using von Frey filaments) in the hind paw for several weeks. The study was performed according to a blind protocol, in which the investigator did not know the identity of the compound/control or the treatment of the animal.
Antibody G2 (anti-CGRP monoclonal antibody) reversed the insufficient load bearing of OA hind limbs at the highest tested dose (10mg/kg) administered IV. The effect is comparable to celecoxib. Both compounds reversed the weight bearing excursion on the OA limb to normal. No effect was observed for the lower doses. This effect lasted 10 days after administration, when the plasma exposure of antibody G2 reached an average of 65.1 ± 3ug/ml (10 days after IV administration, 6.3 ± 0.3ug/ml for lower doses).
The negative control (null antibody, i.e. not binding CGRP) (see figure 1) showed an effect on load bearing at early time points. This effect was unexpected and unexplained, but it was not persistent and was not observed at the allodynic end point (figure 1).
Repeated experiments performed under the same conditions using buffer as a negative control showed no effect on load bearing for the negative control, whereas the shift in load bearing on the OA limb was reversed to normal for G1.
Example 2: binding assays
Binding assays were performed to measure the IC of anti-CGRP antibodies G2 and G1 blocking the binding of human alpha-CGRP to CGRP 1-receptor in SK-N-MC cells50. Dose response curves were plotted using the formula: ki=IC50V. (1+ ([ ligand ]]/KD) Determination of KiA value; FIG. 2, wherein the equilibrium dissociation constant K of human α -CGRP for CGRP 1-receptor present in SK-N-MC cellsD0.37 nM. IC to be reported50The value (in the case of IgG molecules) is converted into a binding site so that it can be determined by Biacore with affinity (K)D) In contrast, N-biotinylated human and rat α -CGRP were captured at low levels (typically 100 response units) on separate flow cells to provide reaction surfaces, while the unmodified flow cell served as a reference channel. G1 was titrated onto the chip surface. Quotient (K) from kinetic rate constantsD=koff/kon) Deducing the binding affinity. See table 1.
| G2 | Mouse Mab7E9 | G1 | |
| KD(nM),α-Hu | 17 | 1.0 | 0.04 |
| IC50(nM)α-Hu | 37 | 2.6 | 1.2 |
| KD (nM) alpha-rat | 1.0 | 58 | 1.2 |
TABLE 1
N-growth on chip
biotin-CGRP.degree.C.kon(1/Ms) koff(1/s) T1/2(h) KD(nM)
TABLE 2
G1 has equal binding affinity for human α and β CGRP (Kd ═ 0.155 and 0.152nM, respectively). G2 bound equally well (16 and 17nM, respectively) to rat α and β CGRP. Furthermore, G1 binding affinity in humans is 40 times that of rats for α -CGRP (Kd 0.042 and 1.22nM, respectively), and G1 binding affinity is equivalent in humans and rats (Kd 0.155 and 0.152nM, respectively) for β -CGRP. Antibody G1 also showed good cross-species selectivity and bound rat α -CGRP with the same affinity as antibody G2 (approximately 1.2nM) (table 2).
G1 binds with high affinity to human and cynomolgus alpha-and beta-CGRP (K, respectively)D63 and 155 pM). G1 demonstrates species selectivity for human/cynomolgus CGRP and binds with lower affinity to alpha-and beta-CGRP (K respectively) from other species such as ratD2.57nM and 152 pM).
Example 3: in vivo half-life of anti-CGRP
Serum measurements of anti-CGRP in rats (figure 3) showed a half-life of about 7 days. With a molecular weight of approximately 150,000, the antibody is restricted to the periphery, fig. 3a, 3b, i.e. it cannot enter the central nervous system or cross the blood-brain barrier.
Example 4: selectivity of anti-CGRP antibodies
We determined the specificity of antibody G1 for human or rat CGRP by "probing" the free concentration of the pre-mixed complexes of mAb + peptides using a Biacore chip. As expected, when we preincubated antibody G1 with human or rat CGRP, the response was completely blocked. In contrast, preincubation of G1 with excess amylin (amylin), calcitonin, or adrenomedullin was comparable to the control response (G1 plus buffer), indicating that G1 does not form complexes with these peptides (fig. 5).
Example 5: identification of binding epitopes of antibody G1
Biacore 3000 on a sensor chip equipped with a streptavidin coating (SA) using standard Biacore running buffer (HBS-P)TMThe interaction analysis was performed on the system (Biacore AB, Uppsala, Sweden) at 25 ℃. First we confirmed that the N-biotinylated 25-37 human α -CGRP fragment binds antibody G1 with the same affinity as full-length N-biotinylated human α -CGRP. The amino acids between positions 27-37 were then mutated individually to alanine and the fold loss of affinity compared to the wild type fragment was detected. The N-biotinylated fragments are captured at low levels (typically 100 response units) on a separate flow cell to provide a reaction surface, while the unmodified flow cell serves as a reference channel. Purified Fab fragments of antibody G1 were generated. The Fab fragments were titrated on-chip using 1 μ M as the highest concentration for the 2-fold dilution series. The bound and the dissociated phases were monitored at 100. mu.l/min for 1 min and 5 min, respectively. The surface was regenerated using a mixture of 35% ethanol +25mM NaOH +0.5M NaCl.
Alanine scan results showed that antibody G1 binds to the C-terminal region of human α -CGRP, particularly residues 25 to 37, and shows specific binding to a region that can be defined as an epitope and is located in the last 5C-terminal amino acids (i.e., G33A to F37A) (i.e., the loss of affinity is significantly increased when the specific binding region is mutated). The G33A and F37A mutations caused the greatest change in affinity (fig. 4). The C-terminal Phe is very important for the selectivity of antibody G1 for CGRP (compare related peptides and gene family members) (fig. 6).
Accordingly, in one embodiment, the invention provides anti-CGRP antagonist antibodies that specifically bind to an epitope defined by amino acids G33 through F37 of CGRP. The anti-CGRP antagonist antibody can specifically bind to an epitope defined by the amino acid sequence GSKAF, more particularly to an epitope of CGRP defined as gxxf, where X can be any amino acid and G33 and F37 are the most important epitope residues (for determining high affinity binding of the anti-CGRP antagonist antibody).
Example 6: analysis of indicators of physical-embedded psychological dependence
Neither antibody G1 nor antibody G2 showed CNS penetration. Furthermore, long-term observations of animals (rats) receiving either antibody at the levels used in the previous examples did not show adverse CNS events such as sedation or excitability/euphoric behaviour compared to control animals. These observations indicate that there is no dependency risk for the antibody, showing a significantly improved safety of the antibody (over existing opiates used in current pain treatments).
Example 7: analysis of indicators of gastrointestinal adverse effects
The 1 month in vivo rat study with antibody G2 and the 1 week comparative study with antibody G1 showed that no adverse gastrointestinal effects were observed in behavior, food intake, stool production, or histopathology compared to control animals. These observations show that there is no gastrointestinal risk for the antibody, thereby showing significantly improved safety of the antibody (over existing NSAIDs used in current pain therapy).
Example 8; g1 and G2 as anti-CGRP antagonist antibodies
A known consequence of CGRP biological activity is the production of neurogenic flushing when delivered in vivo. G1 and G2 proved to be anti-CGRP antagonist antibodies because they prevented the development of neurogenic flushing in vivo.
The efficacy of G1 was tested for the ability of G1 to block the effect of CGRP in vivo by using the neurogenic cutaneous flushing rat model. The saphenous nerve in rats is electrically stimulated, causing release of CGRP from the nerve endings and resulting vasodilation, and the resulting change in blood flow can be measured using the laser doppler method.
The change in blood flow parameter is expressed as the area under the curve (AUC, change in arbitrary doppler flux units times time). CGRP receptor antagonist CGRP8-37(400nmol/kg, i.v.) was used as a positive control to verify the specificity of the model (data not shown). To determine the effect of G1 on each animal prior to dosing, a baseline blood flow response to stimulation was established with two saphenous nerve stimulations (each 30 min apart). After the blood flow response of the second stimulation had returned to baseline levels (approximately 10 minutes post stimulation), the rats were treated with G1 and an additional 4 stimulations were performed at 30 minute intervals.
The results (fig. 7) show no significant change in blood flow response in vehicle-treated animals, but rats treated with G1 showed a significant decrease in blood flow response (for treatments of 10mg/kg and 1mg/kg, starting at 90 and 120 minutes post-dose, respectively). Similar activity was obtained using D2. Furthermore, in additional neurogenic flushing and vasodilation model tests, G1 showed a significant effect 7 days after I V dosing (predicted ED in the saphenous nerve stimulation model506 ug/ml). The conclusion from the tests performed was that G1 and G2 showed anti-CGRP antagonist activity.
Also in publications, Zeller J et al Br J pharmacol.2008 Dec; 155(7): similar CGRP function blocking ability of the antibodies is shown in Epub 2008 Sep 8, 1093-103.
Antibody sequences useful in the practice of the present invention are provided below.
Antibody sequences
Antibody G1 heavy chain variable region amino acid sequence (SEQ ID NO: 1)
EVQLVESGGGLVQPGGSLRLSCAASGFTFSNYWISWVRQAPGKGLEWVAEIRSESDA
SATHYAEAVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCLAYFDYGLAIQNYWGQG
TLVTVSS
Variable region amino acid sequence (S) of antibody G1 light chainEQ ID NO:2)
EIVLTQSPATLSLSPGERATLSCKASKRVTTYVSWYQQKPGQAPRLLIYGASNRYLGIP
ARFSGSGSGTDFTLTISSLEPEDFAVYYCSQSYNYPYTFGQGTKLEIK
Antibody G1 CDR H1 (extended CDR) (SEQ ID NO: 3)
GFTFSNYWIS
Antibody G1 CDR H2 (extended CDR) (SEQ ID NO: 4)
EIRSESDASATHYAEAVKG
Antibody G1 CDR H3(SEQ ID NO: 5)
YFDYGLAIQNY
Antibody G1 CDR L1(SEQ ID NO: 6)
KASKRVTTYVS
Antibody G1 CDR L2(SEQ ID NO: 7)
GASNRYL
Antibody G1 CDR L3(SEQ ID NO: 8)
SQSYNYPYT
Antibody G1 heavy chain variable region nucleotide sequence (SEQ ID NO: 9)
GAAGTTCAGCTGGTTGAATCCGGTGGTGGTCTGGTTCAGCCAGGTGGTTCCCTGC
GTCTGTCCTGCGCTGCTTCCGGTTTCACCTTCTCCAACTACTGGATCTCCTGGGTT
CGTCAGGCTCCTGGTAAAGGTCTGGAATGGGTTGCTGAAATCCGTTCCGAATCCG
ACGCGTCCGCTACCCATTACGCTGAAGCTGTTAAAGGTCGTTTCACCATCTCCCGT
GACAACGCTAAGAACTCCCTGTACCTGCAGATGAACTCCCTGCGTGCTGAAGACA
CCGCTGTTTACTACTGCCTGGCTTACTTTGACTACGGTCTGGCTATCCAGAACTAC
TGGGGTCAGGGTACCCTGGTTACCGTTTCCTCC
Variable region nucleotide sequence of antibody G1 light chain (SEQ ID NO: 10)
GAAATCGTTCTGACCCAGTCCCCGGCTACCCTGTCCCTGTCCCCAGGTGAACGTGCT
ACCCTGTCCTGCAAAGCTTCCAAACGGGTTACCACCTACGTTTCCTGGTACCAGCAGA
AACCCGGTCAGGCTCCTCGTCTGCTGATCTACGGTGCTTCCAACCGTTACCTCGGTAT
CCCAGCTCGTTTCTCCGGTTCCGGTTCCGGTACCGACTTCACCCTGACCATCTCCTCC
CTGGAACCCGAAGACTTCGCTGTTTACTACTGCAGTCAGTCCTACAACTACCCCTACA
CCTTCGGTCAGGGTACCAAACTGGAAATCAAA
Antibody G1 heavy chain full length antibody amino acid sequence (including modified IgG2 as described herein)
(SEQ ID NO:11)
EVQLVESGGGLVQPGGSLRLSCAASGFTFSNYWISWVRQAPGKGLEWVAEIRSESDA
SATHYAEAVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCLAYFDYGLAIQNYWGQG
TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVH
TFPAVLQSSGLYSLSSVVTVPSSN FGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPC
PAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAK
TKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPSSIEKTISKTKGQPREP
QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS
FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
Antibody G1 light chain full length antibody amino acid sequence (SEQ ID NO: 12)
EIVLTQSPATLSLSPGERATLSCKASKRVTTYVSWYQQKPGQAPRLLIYGASNRYLGIP
ARFSGSGSGTDFTLTISSLEPEDFAVYYCSQSYNYPYTFGQGTKLEIKRTVAAPSVFIF
PPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYS
LSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
Antibody G1 heavy chain full length antibody nucleotide sequence (including modified IgG2 as described herein)
(SEQ ID NO:13)
GAAGTTCAGCTGGTTGAATCCGGTGGTGGTCTGGTTCAGCCAGGTGGTTCCCTGC
GTCTGTCCTGCGCTGCTTCCGGTTTCACCTTCTCCAACTACTGGATCTCCTGGGTT
CGTCAGGCTCCTGGTAAAGGTCTGGAATGGGTTGCTGAAATCCGTTCCGAATCCG
ACGCGTCCGCTACCCATTACGCTGAAGCTGTTAAAGGTCGTTTCACCATCTCCCGT
GACAACGCTAAGAACTCCCTGTACCTGCAGATGAACTCCCTGCGTGCTGAAGACA
CCGCTGTTTACTACTGCCTGGCTTACTTTGACTACGGTCTGGCTATCCAGAACTAC
TGGGGTCAGGGTACCCTGGTTACCGTTTCCTCCGCCTCCACCAAGGGCCCATCTG
TCTTCCCACTGGCCCCATGCTCCCGCAGCACCTCCGAGAGCACAGCCGCCCTGG
GCTGCCTGGTCAAGGACTACTTCCCAGAACCTGTGACCGTGTCCTGGAACTCTGG
CGCTCTGACCAGCGGCGTGCACACCTTCCCAGCTGTCCTGCAGTCCTCAGGTCTC
TACTCCCTCAGCAGCGTGGTGACCGTGCCATCCAGCAACTTCGGCACCCAGACCT
ACACCTGCAACGTAGATCACAAGCCAAGCAACACCAAGGTCGACAAGACCGTGGA
GAGAAAGTGTTGTGTGGAGTGTCCACCTTGTCCAGCCCCTCCAGTGGCCGGACCA
TCCGTGTTCCTGTTCCCTCCAAAGCCAAAGGACACCCTGATGATCTCCAGAACCCC
AGAGGTGACCTGTGTGGTGGTGGACGTGTCCCACGAGGACCCAGAGGTGCAGTT
CAACTGGTATGTGGACGGAGTGGAGGTGCACAACGCCAAGACCAAGCCAAGAGA
GGAGCAGTTCAACTCCACCTTCAGAGTGGTGAGCGTGCTGACCGTGGTGCACCAG
GACTGGCTGAACGGAAAGGAGTATAAGTGTAAGGTGTCCAACAAGGGACTGCCAT
CCAGCATCGAGAAGACCATCTCCAAGACCAAGGGACAGCCAAGAGAGCCACAGGT
GTATACCCTGCCCCCATCCAGAGAGGAGATGACCAAGAACCAGGTGTCCCTGACC
TGTCTGGTGAAGGGATTCTATCCATCCGACATCGCCGTGGAGTGGGAGTCCAACG
GACAGCCAGAGAACAACTATAAGACCACCCCTCCAATGCTGGACTCCGACGGATC
CTTCTTCCTGTATTCCAAGCTGACCGTGGACAAGTCCAGATGGCAGCAGGGAAAC
GTGTTCTCTTGTTCCGTGATGCACGAGGCCCTGCACAACCACTATACCCAGAAGAG
CCTGTCCCTGTCTCCAG GAAAGTAA
Full-length antibody nucleotide sequence of antibody G1 light chain (SEQ ID N0: 14)
GAAATCGTTCTGACCCAGTCCCCGGCTACCCTGTCCCTGTCCCCAGGTGAACGTG
CTACCCTGTCCTGCAAAGCTTCCAAACGGGTTACCACCTACGTTTCCTGGTACCAG
CAGAAACCCGGTCAGGCTCCTCGTCTGCTGATCTACGGTGCTTCCAACCGTTACC
TCGGTATCCCAGCTCGTTTCTCCGGTTCCGGTTCCGGTACCGACTTCACCCTGAC
CATCTCCTCCCTGGAACCCGAAGACTTCGCTGTTTACTACTGCAGTCAGTCCTACA
ACTACCCCTACACCTTCGGTCAGGGTACCAAACTGGAAATCAAACGCACTGTGGCT
GCACCATCTGTCTTCATCTTCCCTCCATCTGATGAGCAGTTGAAATCCGGAACTGC
CTCTGTTGTGTGCCTGCTGAATAACTTCTATCCGCGCGAGGCCAAAGTACAGTGGA
AGGTGGATAACGCCCTCCAATCCGGTAACTCCCAGGAGAGTGTCACAGAGCAGGA
CAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACCCTGAGCAAAGCAGAC
TACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGTTCTC
CAGTCACAAAGAGCTTCAACCGCGGTGAGTGCTAA
Comparison of the amino acid sequences of human and rat CGRP (human α -CGRP (SEQ ID NO: 15); human β -CGRP (SEQ ID NO: 16); rat α -CGRP (SEQ ID NO: 17); and rat β -CGRP (SEQ ID NO: 18)):
NH2-ACDTATCVTHRLAGLLSRSGGVVKNNFVPTNVGSKAF-CONH2(SEQ ID NO:15)
NH2-ACNTATCVTHRLAGLLSRSGGMVKSNFVPTNVGSKAF-CONH2(SEQ ID NO:16)
NH2-SCNTATCVTHRLAGLLSRSGGVVKDNFVPTNVGSEAF-CONH2(SEQ ID NO:17)
NH2-SCNTATCVTHRLAGLLSRSGGVVKDNFVPTNVGSKAF-CONH2(SEQ ID NO:18)
antibody G2 heavy chain variable region amino acid sequence (SEQ ID NO: 19)
EVQLQQSGPELVKPGASVKMSCKASGYTFTSSVMHWVKQKPGQGLEWIGYINPYND
GTKYNEKFKGKATLTSDKSSSTAYMELSSLTSEDSAVYYCAKGGNDGYWGQGTTLTV
SS
Antibody G2 light chain variable region amino acid sequence (SEQ ID NO: 20)
EIVLTQSPTTMAASPGEKITITCSASSSISSIYLHWYQQKPGFSPKVLIYRASNLASGVPA
RFSGSGSGTSYSLTIGTMEAEDVATYYCQQGSTIPFTFGSGTKLEIK
Antibody G2 CDR H1 (extended CDR) (SEQ ID NO: 21)
SSVMH
Antibody G2 CDR H2 (extended CDR) (SEQ ID NO: 22)
YINPYNDGTKYNEKFKG
Antibody G2 CDR H3(SEQ ID NO: 23)
GGNDGY
Antibody G2 CDR L1(SEQ ID NO: 24)
SASSSISSIYLH
Antibody G2 CDR L2(SEQ ID NO: 25)
RASNLAS
Antibody G2 CDR L3(SEQ ID NO: 26)
QQGSTIPFT
Antibody G2 heavy chain variable region nucleotide sequence (SEQ ID NO: 27)
GAGGTCCAGCTGCAGCAGTCTGGACCTGAGCTGGTAAAGCCTGGGGCTTCAGTGAAG
ATGTCCTGCAAGGCTTCTGGATACACATTCACTAGCTCTGTTATGCACTGGGTGAAGC
AGAAGCCTGGGCAGGGCCTTGAGTGGATTGGATATATTAATCCTTACAATGATGGTAC
TAAGTACAATGAGAAGTTCAAAGG CAAGGCCACACTGACTTCAGACAAATCCTCCAGC
ACAGCCTACATGGAACTCAGCAGCCTGACCTCTGAGGACTCTGCGGTCTATTACTGTG
CAAAAGGGGGTAACGATGGCTACTGGGGCCAAGGCACTACTCTCACAGTCTCCTCA
Variable region nucleotide sequence of antibody G2 light chain (SEQ ID NO: 28)
GAAATTGTGCTCACCCAGTCTCCAACCACCATGGCTGCATCTCCCGGGGAGAAGATC
ACTATCACCTGTAGTGCCAGCTCAAGTATAAGTTCCATTTACTTGCATTGGTATCAGCA
GAAGCCAGGATTCTCCCCTAAAGTCTTGATTTATAGGGCATCCAATCTGGCTTCTGGA
GTCCCAGCTCGCTTCAGTGGCAGTGGGTCTGGGACCTCTTACTCTCTCACAATTGGCA
CCATGGAGGCTGAAGATGTTGCCACTTACTACTGCCAGCAGGGTAGTACTATACCATT
CACGTTCGGCTCGGGGACAAAGTTGGAAATAAAA
Antibody G2 heavy chain full length antibody amino acid sequence (excluding Fc domain) (SEQ ID NO: 29)
EVQLQQSGPELVKPGASVKMSCKASGYTFTSSVMHWVKQKPGQGLEWIGYINPYNDGT
KYNEKFKGKATLTSDKSSSTAYMELSSLTSEDSAVYYCAKGGNDGYWGQGTTLTVSSAK
TTPPSVYPLAPGSAAQTNSMVTLGCLVKGYFPEPVTVTWNSGSLSSGVHTFPAVLQSDLY
TLSSSVTVPSSTWPSETVTCNVAHPASSTKVDKKIVPRD
Antibody G2 light chain full length antibody amino acid sequence (SEQ ID NO: 30)
EIVLTQSPTTMAASPGEKITITCSASSSISSIYLHWYQQKPGFSPKVLIYRASNLASGVPARF
SGSGSGTSYSLTIGTMEAEDVATYYCQQGSTIPFTFGSGTKLEIKRADAAPTVSIFPPSSEQ
LTSGGASVVCFLNNFYPRDINVKWKIDGSERQNGVLNSWTDQDSKDSTYSMSSTLTLTKD
EYERHNSYTCEATHKTSTSPIVKSFNRNEC
Antibody G2 heavy chain full length antibody nucleotide sequence (excluding Fc domain) (SEQ ID NO: 31)
GAGGTCCAGCTGCAGCAGTCTGGACCTGAGCTGGTAAAGCCTGGGGCTTCAGTGAAG
ATGTCCTGCAAGGCTTCTGGATACACATTCACTAGCTCTGTTATGCACTGGGTGAAGC
AGAAGCCTGGGCAGGGCCTTGAGTGGATTGGATATATTAATCCTTACAATGATGGTAC
TAAGTACAATGAGAAGTTCAAAGGCAAGGCCACACTGACTTCAGACAAATCCTCCAGC
ACAGCCTACATGGAACTCAGCAGCCTGACCTCTGAGGACTCTGCGGTCTATTACTGTG
CAAAAGGGGGTAACGATGGCTACTGGGGCCAAGGCACTACTCTCACAGTCTCCTCAG
CCAAAACGACACCCCCATCTGTCTATCCACTGGCCCCTGGATCTGCTGCCCAAACTAA
CTCCATGGTGACCCTGGGATGCCTGGTCAAGGGCTATTTCCCTGAGCCAGTGACAGT
GACCTGGAACTCTGGATCCCTGTCCAGCGGTGTGCACACCTTCCCAGCTGTCCTGCA
GTCTGACCTCTACACTCTGAGCAGCTCAGTGACTGTCCCCTCCAGCACCTGGCCCAG
CGAGACCGTCACCTGCAACGTTGCCCACCCGGCCAGCAGCACCAAGGTGGACAAGA
AAATTGTGCCCAGGGAT
Full-length antibody nucleotide sequence of antibody G2 light chain (SEQ ID N)O:32)
GAAATTGTGCTCACCCAGTCTCCAACCACCATGGCTGCATCTCCCGGGGAGAAGA
TCACTATCACCTGTAGTGCCAGCTCAAGTATAAGTTCCATTTACTTGCATTGGTATC
AGCAGAAGCCAGGATTCTCCCCTAAAGTCTTGATTTATAGGGCATCCAATCTGGCT
TCTGGAGTCCCAGCTCGCTTCAGTGGCAGTGGGTCTGGGACCTCTTACTCTCTCA
CAATTGGCACCATGGAGGCTGAAGATGTTGCCACTTACTACTGCCAGCAGGGTAG
TACTATACCATTCACGTTCGGCTCGGGGACAAAGTTGGAAATAAAACGGGCTGATG
CTGCACCAACTGTATCCATCTTCCCACCATCCAGTGAGCAGTTAACATCTGGAGGT
GCCTCAGTCGTGTGCTTCTTGAACAACTTCTACCCCAGAGACATCAATGTCAAGTG
GAAGATTGATGGCAGTGAACGACAAAATGGTGTCCTGAACAGTTGGACTGATCAG
GACAGCAAAGACAGCACCTACAGCATGAGCAGCACCCTCACATTGACCAAGGACG
AGTATGAACGACATAACAGCTATACCTGTGAGGCCACTCACAAGACATCAACTTCA
CCCATCGTCAAGAGCTTCAACAGGAATGAGTGTTAA
Claims (31)
1. Use of an anti-CGRP antagonist antibody for the manufacture of a medicament for the prevention and/or treatment of inflammatory pain and/or symptoms of inflammatory pain.
2. The use of claim 1, wherein the medicament is prepared for peripheral administration.
3. The use of claim 1, wherein the medicament is administered peripherally.
4. The use of any one of claims 1 to 3, wherein the medicament is prepared for oral, sublingual, by inhalation, transdermal, subcutaneous, intravenous, intra-arterial, intra-articular, periarticular, local and/or intramuscular administration.
5. The use of claim 4, wherein the medicament is prepared for subcutaneous or intravenous administration.
6. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody acts peripherally after administration.
7. Use according to any one of the preceding claims, wherein the inflammatory pain is arthritic pain.
8. The use of claim 7, wherein the arthritic pain is osteoarthritis pain.
9. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody:
(a) combining CGRP;
(b) blocking CGRP binding to its receptor;
(c) block or reduce activation of CGRP receptors;
(d) inhibiting, blocking, suppressing or reducing the biological activity of CGRP;
(e) increasing CGRP clearance; and/or
(g) Inhibiting the synthesis, production or release of CGRP.
10. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody:
(i) is a human antibody which is capable of,
(ii) is a humanized antibody which is a human antibody,
(iii) is a monoclonal antibody which is capable of binding to a human,
(iv) binds CGRP with a Kd of 50nM or less (as measured by surface plasmon resonance at 37 ℃); and/or
(v) Has an in vivo half-life of at least 7 days.
11. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody specifically binds to the C-terminal region of CGRP.
12. Use according to claim 10, wherein the anti-CGRP antagonist antibody specifically recognizes an epitope defined by the sequence GSKAF.
13. Use according to any one of the preceding claims, wherein the anti-CGRP antibody comprises an amino acid sequence that differs from SEQ ID NO: 1 or 19 VH domains that are at least 90% identical.
14. Use according to any one of the preceding claims, wherein the anti-CGRP antibody comprises an amino acid sequence that differs from SEQ ID NO: 2 or 20 VL domains that are at least 90% identical.
15. The use of claim 14, wherein the anti-CGRP antibody further comprises an amino acid sequence that differs from SEQ ID NO: 1 or 19 VH domains that are at least 90% identical.
16. The use of any one of claims 1 to 12, wherein the anti-CGRP antibody comprises at least one CDR selected from the group consisting of:
(a) as set forth in SEQ ID NO: 3 or 21, CDR H1;
(b) as set forth in SEQ ID NO: 4 or 22, CDR H2;
(c) as set forth in SEQ ID NO: CDR H3 shown in 5 or 23;
(d) as set forth in SEQ ID NO: CDR L1 shown in FIG. 6 or 24;
(e) as shown in SEQ ID NO: CDR L2 shown in 7 or 25;
(f) as set forth in SEQ ID NO: CDR L3 shown in 8 or 26; and
(g) variants of L1, L2 and H2.
17. The use of any one of claims 1 to 12, wherein the anti-CGRP antibody comprises an amino acid sequence that differs from SEQ ID NO: 1 and a VH domain at least 90% identical in amino acid sequence to SEQ ID NO: 2 a VL domain that is at least 90% identical.
18. The use of any one of claims 1 to 12, wherein the anti-CGRP antibody comprises a heavy chain produced by the expression vector with ATCC accession No. PTA-6867.
19. The use of any one of claims 1 to 12, wherein the anti-CGRP antibody comprises a light chain produced by the expression vector with ATCC accession No. PTA-6866.
20. Use according to any one of claims 1 to 12, wherein the anti-CGRP antibody is produced by an expression vector with ATCC accession nos. PTA-6867 and PTA-6866.
21. The use of any one of the preceding claims, wherein the medicament is prepared for peripheral administration by subcutaneous or intravenous injection between 1, 2, 3 or 4 times per month.
22. The use of any one of the preceding claims, wherein the medicament is prepared for peripheral administration at an antibody concentration of 5 to 100 mg/ml.
23. Use according to any one of the preceding claims, wherein the medicament is prepared for peripheral administration at an antibody concentration of 1 to 100mg/kg body weight.
24. The use of any one of the preceding claims, wherein the medicament does not produce CNS impairment of motor coordination or attention.
25. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody is not administered by the central, spinal or intrathecal route.
26. The use of any one of the preceding claims, wherein the anti-CGRP antagonist antibody is not a central, spinal or intrathecal osmolyte molecule.
27. Use according to any one of the preceding claims, wherein the anti-CGRP antibody is combined with one or more additional pharmaceutically active compounds and administered separately, sequentially or simultaneously.
28. The use of claim 27, wherein the one or more additional pharmaceutically active compounds are selected from the group consisting of:
(i) opioid analgesics such as morphine, heroin, hydromorphone, oxymorphone, levorphanol, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, dextropropoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine, or pentazocine;
(ii) non-steroidal anti-inflammatory drugs (NSAIDs) such as aspirin, diclofenac, diflunisal, etodolac, fenbufen, fenoprofen, flufenisal, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamic acid, mefenamic acid, nabumetone, naproxen, oxaprozin, phenylbutazone, piroxicam, sulindac, tolmetin, or zomepirac, or a pharmaceutically acceptable salt thereof;
(iii) barbiturates, such as amobarbital, alprenbital, sec-barbital, butabarbital, mephobarbital, methamphetal, methobarbital, pentobarbital, phenobarbital, secobarbital, talbarbital, thiopentobarbital or thiopental, or pharmaceutically acceptable salts thereof;
(iv) benzodiazepines with sedative effectSuch as chlordiazepoxide, chlordiazepoxideDiazepam, flurazepam, lorazepam, oxazepam, temazepam or triazolam or a pharmaceutically acceptable salt thereof;
(v) h1 antagonists with sedative effects, such as diphenhydramine, mepyramine, promethazine, chlorphenamine or clocycline or pharmaceutically acceptable salts thereof;
(vi) sedatives such as glutethimide, meprobamate, methaqualone or chloralbyrin or pharmaceutically acceptable salts thereof;
(vii) skeletal muscle relaxants, such as baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine, methocarbamol, or oxyphennara, or pharmaceutically acceptable salts thereof;
(viii) NMDA receptor antagonists, such as dextromethorphan ((+) -3-hydroxy-N-methyl morphinan) or its metabolite dextrorphan ((+) -3-hydroxy-N-methyl morphinan), ketamine, memantine, pyrroloquinoline quinone, or cis-4- (phosphonomethyl) -2-piperidinecarboxylic acid, or a pharmaceutically acceptable salt thereof;
(ix) α -adrenergic agents, such as doxazosin, tamsulosin, clonidine or 4-amino-6, 7-dimethoxy-2- (5-methanesulfonamido-1, 2, 3, 4-tetrahydroisoquinol-2-yl) -5- (2-pyridyl) quinazoline;
(x) Tricyclic antidepressants, such as desipramine, imipramine, amitriptyline or nortriptyline;
(xi) Anticonvulsants, such as carbamazepine or valproate;
(xii) Tachykinin (NK) antagonists, in particular NK-3, NK-2 or NK-1 antagonists, such as (alpha R, 9R) -7- [3, 5-bis (trifluoromethyl) benzyl ] -8, 9, 10, 11-tetrahydro-9-methyl-5- (4-tolyl) -7H- [1, 4] diazocino [2, 1-g ] [1, 7] naphthyridine-6-13-dione (TAK-637), 5- [ [ (2R, 3S) -2- [ (1R) -1- [3, 5-bis (trifluoromethyl) phenyl ] ethoxy-3- (4-fluorophenyl) -4-morpholinyl ] methyl ] -1, 2-dihydro-3H-1, 2, 4-triazol-3-one (MK-869), lanopiptan, dapitaptan or 3- [ [ 2-methoxy-5- (trifluoromethoxy) phenyl ] methylamino ] -2-phenyl-piperidine (2S, 3S);
(xiii) Muscarinic antagonists such as oxybutynin, tolterodine, propiverine, trospium chloride or darifenacin;
(xiv) COX-2 inhibitors, such as celecoxib, rofecoxib, or valdecoxib;
(xv) Non-selective COX inhibitors (preferably with GI protection), such as nitroflurbiprofen (HCT-1026);
(xvi) Coal tar analgesics, particularly acetaminophen;
(xvii) Neuroleptic agents such as droperidol;
(xviii) A vanilloid receptor agonist (e.g., resiniferatoxin) or antagonist (e.g., capsaicinoid);
(xix) Beta-adrenergic agents such as propranolol;
(xx) Local anesthetics, such as mexiletine;
(xxi) Corticosteroids, such as dexamethasone;
(xxii) A 5-hydroxytryptamine receptor agonist or antagonist;
(xxiii) A cholinergic (nicotinic) analgesic;
(xxiv) Tramadol (trade mark);
(xxv) PDEV inhibitors, such as sildenafil, vardenafil or tadalafil;
(xxvi) Alpha-2-delta ligands such as gabapentin or pregabalin; and
(xxvii) Cannabinoids are provided.
29. A method of treating and/or preventing inflammatory pain and/or symptoms of inflammatory pain in an individual, the method comprising peripherally administering to the individual a therapeutically effective amount of an anti-CGRP antagonist antibody.
30. A pharmaceutical composition for treating and/or preventing inflammatory pain and/or symptoms of inflammatory pain in an individual comprising an anti-CGRP antagonist antibody and a pharmaceutically acceptable carrier, wherein the composition is prepared for peripheral administration.
31. A kit, comprising:
(a) the pharmaceutical composition of claim 30, and
(b) instructions for peripherally administering to the subject a therapeutically effective amount of the pharmaceutical composition to treat and/or prevent inflammatory pain and/or symptoms of inflammatory pain.
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US61/033,568 | 2008-03-04 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| HK1148230A true HK1148230A (en) | 2011-09-02 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20220017608A1 (en) | Methods of Treating Inflammatory Pain | |
| US10597448B2 (en) | Methods for treating visceral pain associated with interstitial cystitis by administering antagonist antibodies directed against calcitonin gene-related peptide | |
| HK1245098A1 (en) | Methods of treating chronic pain | |
| HK1233486A1 (en) | Methods of treating inflammatory pain | |
| HK1148230A (en) | Methods of treating inflammatory pain | |
| HK1148453A (en) | Methods of treating chronic pain | |
| HK1177435A (en) | Methods for treating visceral pain by administering antagonist antibodies directed against calcitonin gene-related peptide |