MXPA06005469A - Il-18 binding proteins - Google Patents

Il-18 binding proteins

Info

Publication number
MXPA06005469A
MXPA06005469A MXPA/A/2006/005469A MXPA06005469A MXPA06005469A MX PA06005469 A MXPA06005469 A MX PA06005469A MX PA06005469 A MXPA06005469 A MX PA06005469A MX PA06005469 A MXPA06005469 A MX PA06005469A
Authority
MX
Mexico
Prior art keywords
seq
residues
binding protein
human
antibody
Prior art date
Application number
MXPA/A/2006/005469A
Other languages
Spanish (es)
Inventor
Jia Xiaochi
Ghayur Tariq
Labkovsky Boris
W Voss Jeffrey
Green Larry
Babcook John
Wieler James
Singh Kang Jaspal
Hedberg Brad
Original Assignee
Abbott Laboratories*
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Laboratories* filed Critical Abbott Laboratories*
Publication of MXPA06005469A publication Critical patent/MXPA06005469A/en

Links

Abstract

The present invention encompasses IL-18 binding proteins, particularly antibodies that bind human interleukin-18 (hIL-18). Specifically, the invention relates to antibodies that are entirely human antibodies. Preferred antibodies have high affinity for hIL-18 and/or that neutralize hIL-18 activity in vitro and in vivo. An antibody of the invention can be a full-length antibody or an antigen-binding portion thereof. Method of making and method of using the antibodies of the invention are also provided. The antibodies, or antibody portions, of the invention are useful for detecting hIL-18 and for inhibiting hIL-18 activity, e.g., in a human subject suffering from a disorder in which hIL-18 activity is detrimental.

Description

PROTEINS OF UNION IL-18 Reference to the Related Request This application claims the benefit of priority to the US application. No. 10/706, 689, filed on November 12, 2003.
Field of the Invention The present invention relates to binding proteins of interleukin 1 8 (IL-1 8), and specifically to its uses in the prevention and / or treatment of acute and chronic inflammatory diseases.
Background of the Invention Interleukin 1 8 (IL-18) was originally described in 1989 as a factor inductor of interferon gamma (IGIF) and is a pro-inflammatory cytokine with several functions in addition to an ability to induce interferon gamma. These biological properties include the activation of Fas binder expression, the induction of chemokines both CC and CXC, and the increased production of competent human immunodeficiency virus. Due to the ability of I L-1 8 to induce the production of interferon gamma in T cells and macrophages, it plays an important role in Th1 immune responses and participates in both innate and acquired immunity. For revisions of structure, function and biological activity, see for example, Dinarello, C. ef al. (1998) J. Leukoc S / o? 63: 658-654; Dinarello, C. A. (1999) Methods 19 :; and Dinarello, CA (1999; J. Allergy Clin., 103: 11-24; I. B. et al. (2000) Immunology Today 21: 312-315; Nakanishi, K. et al (2001) Ann. Rev. : Immunol 19: 423-474 Intracellular Pro-IL-18 is proteolytically processed to an active form 18 kDa in cells stimulated by endotoxin by caspase 1 (Ghayur, T. er al., (1997) Nature 386: 619-623; Gu, et al., (1997) Science 275: 206-209) and in cells stimulated by bacterial DNA or Fas-L by caspases 4, 5 and 6 (Tsutsui, H. et al., (1999) 11: 359- 67; Ghayur, T., Unpublished Observations.) Pro-IL-18 is also proteolytically processed by other proteases such as neutrophil proteinase 3 (Sugawara, S. et al., (2001) J. 167.6568-6575), caspase 3 (Akita, K. ef al., (1997) 272.26595-26603), and proteases of serine elastase and cathepsin (Gracie JA, et al., (2003) Journal of Leukocyte Biology 73,213-224). Both human and murine lack a classical leader sequence and the mechanism of IL-18 release by cells is not understood. The biological activities of IL-18 are mediated through the binding of IL-18 to a 1L-18 heterodimeric receptor (IL-18R) consisting of two subunits: the a subunit (a member of the IL-18 family). IR, also called protein 1 related to IL-1R or IL-1Rrp1) and the β subunit (also called accessory protein IL-18R, IL-18AP and AcPL). The IL-18Ra subunit binds directly to IL-18, but is unable to signal transduction. The β subunit does not bind to IL-18 by itself, but together with the subunit or. it forms the high affinity receptor (KD = ~ 0.3 nM) that is required for signal transduction (Sims, J. E., (2002) Current Opin Immunol., 14: 1, 17-122). The signal transduction of IL-18 by means of the IL-1 8Raß complex is similar to the IL-IR and Toll type receptor systems (TLR). IL-18R signaling utilizes signal transduction molecules, such as MyD88, IRAK, TRAF6, and results in similar responses (eg, activation of NI K, kinase I kB, NF-kB, JNK and p38 MAP kinase) as IL-1 does. The requirement for I L-18Ra and signal transduction molecules in mediating the bioactivity of IL-18 has been confirmed using the IL-18Ra subunit (Hoshino K., et al (1999) J. Immunol. 162: 5041- 5044;), (Adachi et al. (1998) Immunity 9: 143-150) or IRAK (Kanakaraj P., (1999) J. Exp. Med. 189: 1 129-1 1 38) deletions respectively. The antibodies that bind IL-18 are known in the art.
Mouse antibodies capable of neutralizing IL-18 are described in EP 0 974 600. Human antibodies to IL-1 8 have been described in PCT application WO 01 58956 and are incorporated herein by reference. The present invention provides a new family of binding proteins, human antibodies, and fragments thereof, IL-18 capable of binding, binding with high affinity, and binding and neutralization of IL-18.
BRIEF DESCRIPTION OF THE INVENTION This invention pertains to IL-1 binding proteins 8, particularly antibodies to human I L-18, as well as methods for making and using such binding proteins. One aspect of the invention pertains to a method for regulating gene expression using an IL-18 modulator. One aspect of this invention pertains to a binding protein comprising an antigen binding domain capable of binding human IL-18. In one embodiment, the antigen-binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of: CDR-H1. X -.- X2-X3-X4-X5-X6-X7 (SEQ ID NO: 42), wherein; X! is S, N, H, R, or Y; X2 is Y, G, R, S, or C; X3 is W, G, Y, D, S, V, or I; X4 is H, W, Y, M, L, or D; X5 is G, Y, S, N, or H; X6 is W, or does not occur; and X7 is T, S, G, or does not occur; CDR-H2. X-] -X2-X3-X -X5-X6-X -X8-X9-X? o-X? ? -X? 2-X-i3-X? - i 5-X .6- .7 (SEQ ID NO: 43), wherein; X2 is or F; X3 is Y, S, or W; X4 is P, Y, or S; X5 is G, S, R, or D; X6 is D, or G; X7 is S, T, G, or R; X8 is E, T, I or N; X9 is T, Y, N, K, or H; X10 is R, Y, or S; Xn is Y, N, or S; X? 2 is S, P, A, or V; X13 is P, S, or D; X 4 is T, L, or S; X15 is F, K, or V; X16 is Q, S, or K; and X17 is G, or does not occur; CDR-H3. X -? - X2-X3-X4-Xs-X6-X7-X8-9"Xl 0-XI 1-XI2-XI 3-XI-X-I5-XI 6" X17-X? ß (SEQ ID NO: 44), wherein; XT is V, D, E, S, or C; X2 is G, R, D, S, K, L, Y, or A; X3 is G, Y, R; X4 is G, S, Y, N, T, or D; X5 is W, S, A, G, Y, or T; X6 is Y, G, S, F, W, or N; X7 is P, S, F, Y, V, G, W, or V; X8 is Y, F, D, P, M, I or N; X9 is T, W, D, L, Y, E, P, F, or G; X10 is F, D, Y, H, V, Y, or does not occur; Xn is D, Y, F, L, or does not occur; X-? 2 is I, D, Y, or does not occur; X-I 3 is Y, or does not occur; X1 is Y, or does not occur; X 5 is G, or does not occur; X16 is M, or does not occur; X17 is D, or does not occur; and X18 is V, or does not occur; CDR-L1. X? -X2-X3-X -X5-X6-X7-X8-X9-? o-X? ? -X? 2-Xi3-X? -Xi5-Xi6-Xi7 (SEQ: 45), wherein; X2 is A, G, or S; X3 is S; X4 is E, R, Q, or H; X5 is S, I, T, or N; X6 is I, V, L, or F; X7 is S, G, L, N, or R; X8 is S, G, Y, R, N, H, or D; X9 is N, G, Y, R, or S; X10 is L, Y, S, or D; X-n is A, L, N, V, G, or D; X? 2 is A, N, E, K, G, or does not occur; X13 is K, T, N, or does not occur; X? 4 is N, Y, T, or does not occur; X? 5 is Y, L, or does not occur; X-i6 is L, C, Y, or does not occur; and X17 is A, D, or does not occur; CDR-L2. X -.- X2-X3-X4-X5-X6-X7 (SEQ NO: 46), wherein; X2 is A, V, T, I, or L; X3 is S, or F; X4 is T, I, N, S, R, or Y; X5 is R, or L; X6 is A, Q, E, or F; and X7 is T, or S; and CDR-L3. (SEQ ID NO: 47), wherein; X2 is Q, H, or Y; X3 is Y, N, G, S, or R; X4 is N, H, Y, D, G, V, L, or I; X5 is G, I, Y, S, Q, F, or E; X6 is W, S, T, L, I, or F; X7 is P, L, T, D, or I; X8 is S, L, P, C, W, I or F; X9 is I, T, S, or does not occur; and X-io is T, or does not occur. Preferably, the antigen binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of Residues 31-35 of SEQ ID NO: 6; Residues 50-66 of SEQ ID NO .: 6; Residues 99-110 of SEQ ID NO .: 6; Residues 24-34 of SEQ ID NO .: 7; Residues 50-56 of SEQ ID NO .: 7; Residues 89-98 of SEQ ID NO .: 7; Residues 31-37 of SEQ ID NO .: 8; Residues 52-67 of SEQ ID NO .: 8; Residues 100-110 of SEQ ID NO .: 8; Residues 24-35 of SEQ ID NO .: 9; Residues 21-27 of SEQ ID NO .: 9; Residues 90-98 of SEQ ID NO .: 9; Residues 31-35 of SEQ ID NO .: 10; Residues 50-65 of SEQ ID NO .: 10; Residues 98-107 of SEQ ID NO .: 10; Residues 24-34 of SEQ ID NO .: 11; Residues 50-56 of SEQ ID NO .: 11; Residues 89-97 of SEQ ID NO .: 11; Residues 31-37 of SEQ ID NO .: 12; Residues 52-67 of SEQ ID NO .: 12; Residues 100-108 of SEQ ID NO .: 12; Residues 24-35 of SEQ ID NO .: 13; Residues 51-57 of SEQ ID NO: 13; Residues 90-98 of: 13; Residues 31-35 of SEQ ID NO .: 14; Residues 50-66 of SEQ ID NO .: 14; Residues 99-111 of SEQ ID NO .: 14; Residues 24-40 of SEQ ID NO .: 15; Residues 56-62 of SEQ ID NO: 15; Residues 95-103 of SEQ ID NO .: 15; Residues 31-37 of SEQ ID NO .: 16; Residues 52-67 of SEQ ID NO .: 16; Residues 100-109 of SEQ ID NO: 16; Residues 24-35 of SEQ ID NO .: 17; Residues 51-57 of SEQ ID NO .: 17; Residues 90-98 of SEQ ID NO .: 17; Residues 31-35 of SEQ ID NO .: 18; Residues 20-36 of SEQ ID NO .: 18; Residues 99-108 of SEQ ID NO .: 18; Residues 24-34 of SEQ ID NO: 19; Residues 50-56 of SEQ ID NO .: 19; Residues 89-97 of SEQ ID NO .: 19; Residues 31-35 of SEQ ID NO: 20; Residues 52-67 of SEQ ID NO: 20; Residues 100-108 of SEQ ID NO .: 20; Residues 24-35 of SEQ ID NO .: 21; Residues 51-57 of SEQ ID NO .: 21; Residues 90-98 of SEQ ID NO .: 21; Residues 31-35 of SEQ ID NO .: 22; Residues 50-66 of SEQ ID NO .: 22; Residues 99-116 of SEQ ID NO .: 22; Residues 24-39 of SEQ ID NO .: 23; Residues 55-61 of SEQ ID NO: 23; Residues 94-102 of SEQ ID NO: 23; Residues 31-37 of SEQ ID NO .: 24; Residues 52-67 of SEQ ID NO .: 24; Residues 100-109 of SEQ ID NO .: 24; Residues 24-35 of SEQ ID NO .: 25; Residues 51-57 of SEQ ID NO .: 25; Residues 90-98 of SEQ ID NO .: 25; Residues 31-37 of SEQ ID NO .: 26; Residues 52-67 of SEQ ID NO .: 26; Residues 100-109 of SEQ ID NO .: 26; Residues 24-35 of SEQ ID NO .: 27; Residues 51-57 of SEQ ID NO: 27; Residues 90-98 of SEQ ID NO .: 27; Residues 31-37 of SEQ ID NO .: 28; Residues 52-67 of SEQ ID NO .: 28; Residues 100-108 of SEQ ID NO .: 28; Residues 24-35 of SEQ ID NO .: 29; Residues 51-57 of SEQ ID NO .: 29; Residues 90-98 of SEQ ID NO .: 29; Residues 31-37 of SEQ ID NO .: 30; Residues 52-67 of SEQ ID NO: 30; Residues 99-109 of SEQ ID NO: 30; Residues 24-35 of SEQ ID NO .: 31; Residues 51-57 of SEQ ID NO: 31; Residues 90-98 of SEQ ID NO .: 31; Residues 31-37 of SEQ ID NO .: 32; Residues 52-67 of SEQ ID NO .: 32; Residues 100-109 of SEQ ID NO .: 32; Residues 24-35 of SEQ ID NO .: 33; Residues 51-57 of SEQ ID NO .: 33; Residues 90-98 of SEQ ID NO .: 33; Residues 31-37 of SEQ ID NO .: 34; Residues 52-67 of SEQ ID NO .: 34; Residues 100-108 of SEQ ID NO .: 34; Residues 24-35 of SEQ ID NO .: 35; Residues 51-57 of SEQ ID NO: 35; Residues 90-98 of SEQ ID NO .: 35; Residues 31-35 of SEQ ID NO .: 36; Residues 50-66 of SEQ ID NO .: 36; Residues 99-116 of SEQ ID NO .: 36; Residues 24-39 of SEQ NO .: 37; Residues 55-61 of SEQ ID NO .: 37; Residues 94-102 of SEQ ID NO .: 37; Residues 31-35 of SEQ ID NO .: 38; Residues 50-66 of SEQ ID NO .: 38; Residues 99-108 of SEQ NO .: 38; Residues 24-35 of SEQ ID NO .: 39; Residues 51-57 of SEQ ID NO .: 39; Residues 90-98 of SEQ ID NO .: 39; Residues 31-37 of SEQ ID NO .: 40; Residues 52-67 of SEQ ID NO .: 40; Residues 97-109 of SEQ ID NO .: 40; Residues 24-40 of SEQ ID NO .: 41; Residues 56-62 of SEQ ID NO .: 41; Residues 95-103 of SEQ ID NO: 41. Preferably, the binding protein comprises at least 3 CDRs. In another preferred embodiment, the binding protein comprises a VH domain. Preferably the VH domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 6; SEQ ID NO: 8; SEQ ID NO: 10; SEQ ID NO: 12; SEQ ID NO: 14; SEQ ID NO: 16; SEQ ID NO: 18; SEQ ID NO: 20; SEQ ID NO: 22; SEQ ID NO: 24; SEQ ID NO: 26; SEQ ID NO: 28; NO: 30; SEQ ID NO: 32; SEQ ID NO: 34; SEQ ID NO: 36; SEQ ID NO: 38; and SEQ ID NO: 40. In another embodiment, the binding protein comprises a VL domain. Preferably the VL domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 7; SEQ ID NO: 9; SEQ NO: 11; SEQ ID NO: 13; SEQ ID NO: 15; SEQ ID NO: 17; SEQ ID NO: 19; SEQ ID NO: 21; SEQ ID NO: 23; NO: 25; SEQ ID NO: 27; SEQ ID NO: 29; SEQ ID NO: 31; SEQ ID NO: 33; SEQ ID NO: 35; SEQ ID NO: 37; SEQ ID NO: 39; and SEQ ID NO: 41. More preferably, the binding protein comprises a VL domain comprising an amino acid sequence of SEQ ID NO: 7 and a VH domain comprising an amino acid sequence of SEQ ID NO: 6.
In another embodiment, the binding protein further comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a constant domain of human IgM; a constant domain of human IgG1; a constant human IgG'2 domain; a constant human IgG3 domain; a constant human IgG4 domain; a human IgE constant domain and a human IgA constant domain. Preferably, the heavy chain immunoglobulin constant region domain is a human IgG 1 constant domain. Preferably, at least one amino acid residue is replaced in the heavy chain constant region domain such that the effector functions of the antibody are altered. More preferably, the human lgG1 constant domain comprises amino acid sequence selected from the group consisting of SEQ ID NO: 2, and SEQ ID NO: 3. In another embodiment, the binding protein further comprises a constant domain of light chain immunoglobulin selected from the group consisting of a constant kappa domain of human Ig; and a constant domain Iambda human Ig. Preferably, the human IgG kappa constant domain comprises amino acid sequence SEQ ID NO: 4 and the constant human lambda Ig domain comprises amino acid sequence SEQ ID NO: 5. In another embodiment the binding protein comprises a constant heavy Ig region having a amino acid sequence selects from the group consisting of: SEQ ID NO: 2, and SEQ ID NO: 3; a constant light region Ig having an amino acid sequence selected from the group consisting of: SEQ ID NO: 4 and SEQ I D NO: 5; a variable heavy region Ig having an amino acid sequence of SEQ ID NO: 6; and a variable Ig light region having an amino acid sequence of SEQ ID NO: 7. In another embodiment, the binding protein comprises a constant heavy region Ig having an amino acid sequence of SEQ ID NO: 3; a constant light region Ig having an amino acid sequence of SEQ ID NO: 4; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 6; and a variable Ig light region having an amino acid sequence of SEQ ID NO: 7. In another embodiment, the linkage is selected from the group consisting of an immunoglobulin molecule or functional variants thereof known in the art, the variants of which are retained the characteristic binding property of the binding protein. Examples of immunoglobulin modalities include but are not limited to a scFv; a monoclonal antibody; a human antibody; a chimeric antibody; a humanized antibody; a single domain antibody; an Fb fragment; a Fab 'fragment; an F (ab ') 2; an Fv; a bound disulfide Fv; and a specific dual or bispecific antibody. More preferably, the binding protein is a human antibody. Another aspect of the invention provides a neutralizing binding protein comprising any of the binding proteins described above wherein the neutralizing binding protein is capable of neutralizing IL-18. Preferably, the neutralizing binding protein is capable of neutralizing any of pro-human IL-18; I L-18 mature-human or I L-1 8 truncated-human. In another embodiment, the neutralizing binding protein decreases the ability of I L-1 8 to bind to its receptor. Preferably, the neutralizing binding protein decreases the ability of pro-human 1L-18; I L-1 8 mature-human or truncated-human IL-18 to bind to its receptor. In another embodiment, the neutralizing binding protein is capable of inhibiting one or more biological activities IL-18 selected from the group consisting of Th1 modulation; Th2 modulation (Nakanishi K., ef a. I (2001) Cytokine and Growth Factor Rev. 12: 53-72); Nk modulation; neutrophil modulation; monocyte-macrophage lineage modulation; neutrophil modulation; eosinophil modulation; B cell modulation; cytokine modulation; chemokine modulation; adhesion molecule modulation; and modulation of cell recruitment. In a preferred embodiment, the neutralizing binding protein has a dissociation constant (KD) selected from the group consisting of: more than about 10/7 M to more than about 10 8 M; to more than about 10 ~ 9 M; to more than about 1 0 ~ 10 M; to more than about 10 0"11 M to more than about 10" 12 M; and to more than approximately 10"13M.
In another embodiment, the neutralizing binding protein has an active scale selected from the group consisting of: at least about 102M "V1, at least about 1 03M" V; at least about 104M "V; at least about 105M" 1S "1; and at least about 1 06M" V1. In yet another embodiment, the neutralizing binding protein has an inactive scale selected from the group consisting of: at more than about 10"3s" 1; to more than about 10"4s" 1; to more than about 10"5s" 1; and more than about 1 0"6s" 1. Another aspect of the invention provides a labeled binding protein comprising any of the binding proteins described above wherein the binding protein is conjugated to a detectable label. Preferably, the detectable label is selected from the group consisting of a radiolabel, an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label and biotin. More preferably, the radiolabel is 3H, 14C, 35S, 90Y, "Te, 1111, 125I, 131L, 177Lu, 166Ho, or 153Sm." Another aspect of the invention provides a conjugated protein comprising any of the binding proteins described above wherein said binding protein is conjugated to a cytotoxic or therapeutic agent Preferably, the cytotoxic or therapeutic agent is selected from the group consisting of an anti-metabolite, an alkylating agent, an antibiotic, a growth factor; cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, toxin, and an apoptotic agent.A modaliad belongs to an isolated nucleic acid encoding any of the binding proteins described above. it comprises the above-described isolated nucleic acid wherein said vector is selected from the group consisting of pcDNA; pTT ((Durocher et al., Nucleic Acids Res. earch 2002, Vol 30, No. 2); pTT3 (pTT with additional multiple cloning site; (Mizushima, S. and Nagata, S .; (1990) Nucleic acids Research Vol 18, No. 17); pBV; pJV; and pBJ In another embodiment, a host cell is transforms with the vector.Preferably, the host cell is a prokaryotic cell.Most preferably, the host cell is E.coli.In a related embodiment, the host cell is a eukaryotic cell.Preferably, the eukaryotic cell is selected from the group consisting of: of protist cell, animal cell, plant cell and fungal cell More preferably, the host cell is a mammalian cell that includes, but is not limited to, CHO and COS, or a fungal cell such as Saccharomyces cervisiae, or a cell insect such as Sf9. Another aspect of the invention provides a method of producing a binding protein that binds human IL-18, comprising culturing the host cells, comprising culturing any of the anti-host cells. described above in a culture medium under conditions sufficient to produce a binding protein that binds human IL-18. Another embodiment provides a binding protein produced according to the method disclosed above. Another aspect of the invention provides a crystallized binding protein comprising any of the mandatory proteins described above, wherein the binding protein exists as a crystal. The crystal is preferably a vehicle controlled pharmaceutical release crystal. In one embodiment the binding protein that exists as a crystal has a longer period in vivo than the soluble counterparts of the binding protein. In another embodiment, the binding protein retains its biological activity after crystallization. One embodiment provides a composition for the release of a binding protein wherein the composition comprises a formulation which in turn comprises a recrystallized binding protein as described above and an ingredient; and at least one polymeric vehicle. Preferably, the polymer carrier is a polymer selected from one or more of the group consisting of; poly (acrylic acid), poly (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-go-glycolic acid) or PLGA, poly (b-hydroxybutyrate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organ) phosphazene); poly (ortho esters), poly (vinyl alcohol), poly (vinylpyrrolidone), vinyl ether-maleic anhydride copolymers, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycolminoglycans, sulfated poly-archaarides, mixtures and copolymers thereof. Preferably the ingredient is selected from the group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hidoxypropyl-β-cyclodextrin, methoxypolyethylene glycol and polyethylene glycol. Another embodiment provides a method for treating a mammal comprising the step of administering to the mammal an effective amount of the composition described above. Another aspect of the invention provides a method for regulating gene expression of a gene of interest comprising the steps of providing an IL-18 polypeptide or a modulated IL-18; and contacting the polypeptide or modulator to a cell wherein the gene of interest is selected from the group consisting of genes identified by GenBank Identification numbers: NM_, 000389- N _002198, NM_002163, NM_006144, NM_006515, NM__0G7185, NML.00 -_. 88, NM_003661, M 021958, NM__001335, Hs.382006, NM_02OI25, NM_007210, NM_021798, NML01332, MI 1313, D88152, NM_001103, U37519, NM_000697, J03600. NM_014578, S66793, U47054, L19871. M81181, NM .001I88, U15460, NML.0144I7, Z23115, NM_001713, U45878, U37546, U72649, U49187, J03507, U50360 XML071866, M_I) 05623, Z32765, Z11697, XM 071866, U51096, M83667, D87469, L07765, U66468, X14830, L29217, X15880, NM .001851, M27691, M37435, X13589, X16866, X59131, N3M.00 393, U73328, LI9267, U53445, X68277, U48807, NMJXH950, U87269, M57730 , X52541, J04076, XB3741, L07077, M62831, M6O830, U53786, NM__0019885 NML.000141, M23668, U60062, MM_000141, U49973, U89995, U27326, A28102, M2S667, L34357. U19523, L01406, U03486, X68285, Z18859, D49958, D43772, AC000099, M57731, X53800, M91036, D16583, X64877, X58431, M16937, NM_014468, X92814, L19314, M26665, D10995, L41147, M24283, S81914, J03171, J00219, - ^ 000619. NMJ0OO5S5, U31628, X04500, M27492, X01057, M26062, Y00081, Y00787, Z31695, X06256, X57206, U20734. NM__014879, D31762, D42038, NM .005551- NM_, 014846, X061S2, M-U.05551. XO7730, M13955, M57710, S83362, NM 002314, NM_00S569, U49957, U89922, X14008, U59914, D14497, X59727, NM_000429, U43944, X72755, NM__Q21230, NM_005951, X78710, X70991, M32011, S77763, M58603, S76638, M69043, U91616 , D86425, L13740, U44848, TJ79251, M27288, AF000234, DS0640, L20971, L10343, U77735, N.003579, U17034, AB000584, X63131, DI 1428, NM B2940, Nlvl_005035, NM_003579, M18255. L01087, D38128, Y10375, D15049, M31166, U59877, NM_003579, U64675, S57153, NM_002903, NGJQ00013, X75042, M83221, NML 000537, U22314, S59049, U70426, U22377, U38480, L10338, M23178, M69203, NM_005409. D79206, N_V-) Q5065, NM..004186, J03764, NM_006802, D89077, NM_003037, M91463, D82326, L05568, U96094, X83301, D21267, L31529, M62800, NM_021014, Z35093, NML.005816, L25444, 95787, NM. 005421, L47345, M57732, NM .003205, M96956, U19878, M92357, M59465, X83490, U37518, NM_003294, U19261, U78798, S69790, U53476, L15309, U78722, X57809, U79249, AB000464, X77744, U79248, AI420129, HG2981- HT3127, HG3548-H 3749, HG870-HT870, HG4333-HT4603, HG3ip-HT3287, HG4593-HT4998, HG961-BT961, HG1877-HT1917, HG3p5-HT3291, HG4115-HT4385, and HG3925-HT4195.
Preferably, the modulator is an antagonist. More preferably the adulator is a binding protein or a neutralizing binding protein. The invention also provides a pharmaceutical composition comprising a binding protein or a neutralizing binding protein. The invention also provides a pharmaceutical composition comprising a binding protein or a neutralizing binding protein as described above and a pharmaceutically acceptable carrier. In a further embodiment, the pharmaceutical composition comprises at least one additional therapeutic agent for treating a disorder in which the activity of IL-18 is deleterious. Preferably, the additional agent is selected from the group consisting of angiogenesis inhibitors (including but not limited to anti-VEGF antibodies or VEGF coatings); kinase inhibitors (including but not limited to inhibitors KDR and TIE-2); co-stimulation molecule blockers (including but not limited to anti-B7.1, and anti-B7.2, CTLA-4-lg, anti-CD20); adhesion molecule blockers (including but not limited to anti-LFA-1 Abs; anti-E / L selectin Abs, small molecule inhibitors); anti-cytokine antibody or functional fragment thereof (including but not limited to anti-I L-12, anti-TNF, anti-IL-6 / cytokine receptor antibodies); methotrexate; corticosteroids; cyclosporin; rapamycin; FK506; and non-inflammatory and non-steroidal agents. In another aspect, the invention provides a method for inhibiting the activity of human IL-1 comprising contacting human IL-1 with a binding protein described above such that the activity of human L-18 is inhibited. In a related aspect, the invention provides a method for inhibiting the activity of human IL-18 in a human subject suffering from a binding protein described above in such a way that the activity of human IL-1 in the human subject is inhibited and the treatment is achieved. Preferably, the disorder is selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropathy, systemic lupus erythematosus, Chron's disease, ulcerative colitis, inflammatory bowel disease , insulin-dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, scleroderma dermatitis, graft against host disease, rejection of organ transplantation (including but not limited to bone marrow and solid organ rejection), chronic or acute immune disease, disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawashi disease, severe disease, nephrotic syndrome, chronic fatigue syndrome, Wegener granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, shock s optical, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington chorea, Parkinson's disease, Alzheimer's disease, primary biliary ciris, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt syndrome, adult respiratory distress syndrome (acute), alopecia, alopecia areata, ulcerative eolithic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy , spondylopathy, atheromatous disease / arteriosclerosis, atopic allergy, autoimmune bolus disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune hemolytic anemia, positive hemolytic anemia Coombs, acquired pernicious anemia, juvenile pernicious anemia, myalgic encephalitis / Royal Free disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosis hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Related Diseases - Acquired Immunodeficiency, Hepatitis B, Hepatitis C, common variable immunodeficiency (common variable hypogammaglobulinaemia) , dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, alveolites of cryptogenic fibrosis, post-inflammatory interstitial lung disease, interstitial pneumonitis, interstitial lung disease associated with connective tissue disease, associated lung disease with mixed connective tissue disease, interstitial lung disease associated with systemic sclerosis, interstitial lung disease associated with rheumatoid arthritis, lung disease associated with systemic lupus erythematosus, lung disease associated with dermatomyositis / polymyositis, lung disease associated with Sjogren disease, lung disease associated with ankylosing spondylitis, diffuse vasculitic lung disease, lung disease associated with hemosiderosis, drug-induced interstitial lung, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, post-infectious interstitial lung disease, gout arthritis, autoimmune hepatitis, autoimmune hepatitis, autoimmune hepatitis type 1 (lupoid or autoimmune hepatitis) classic), autoimmune hepatitis type 2 (anti-LKM antibody hepatitis), autoimmune mediated hypoglycemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute autoimmune disease associated with organ transplantation, chronic immune disease associated with the organ transplantation, osteoarthrosis, primary sclerosis cholangitis, psoriasis type 1, psoriasis type 2, idiomatic leucopaenia, autoimmune neutropenia, NOS renal disease, NOS, glomerulonefritides, microscopic vasculitis of the kidneys, Lyme disease, discoid lupus erythematosus, male or female language infertility NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, syndrome of Sjógren, Takayusu disease / arteritis, autoimmune thrombocytopenia, idiomatic thrombocytopenia, autonimmune thyroid disease, hyperthyroidism, autoimmune hypothyroidism (Alzheimer's disease), autoimmune hypothyroidism atrophy, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo, disease of acute liver, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, coleosatatis, idiosyncratic liver disease, drug-induced hepatitis, nonalcoholic steatohepatitis, allergy and asthma, group B streptococci infection (GBS), mental disorders (for example, depression and schizophrenia), Type Th 1 and Type Th2 mediated diseases, and cancers such as lung, breast, stomach, gallbladder, colon, pancreas, ovary, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma). In another aspect, the invention provides a method for treating a patient suffering from a disorder in which I L-1 8 is deleterious comprising the step of administering any of the binding proteins previously described above, concurrently, or after administration of a second agent, as discussed above. Another aspect of the invention provides a neutralizing binding protein selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and a CDR-grafted antibody, wherein the neutralizing binding protein is capable of I-L-8 mature-human binding, but does not specifically bind pro-human IL-18. Another aspect of the invention provides a neutralizing binding protein selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and an antibody with CDR graft, wherein the neutralizing binding protein is capable of competing with 125-2H antibody to bind human IL-18. Another aspect of the invention provides a neutralizing binding protein selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and a CDR-grafted antibody, wherein the neutralizing binding protein is not capable of competing with 125-2H antibody to bind human L-1 8. Another aspect of the invention provides a neutralizing binding protein selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and a CDR graft antibody, wherein the neutralizing binding protein is not capable of competing with the binding protein selected from the group consisting of 2.5 (E) ml of antibody and IL-1 8BP for I L binding -18 human. In a preferred embodiment, the binding protein is capable of binding mature-human IL-18, but not specifically binding pro-human IL-18. In yet another embodiment, the binding protein is capable of competing with the 125-2H antibody to bind human IL-1. In another embodiment, the binding protein is not able to compete with the 125-2H antibody to bind human IL-18. In yet another embodiment, the binding protein is not capable of competing with a binding protein selected from the group consisting of antibody 2.5 (E) mgl, and IL-18BP to bind human IL-18. In a preferred embodiment, the binding protein comprises a VL domain comprising an amino acid sequence of SEQ ID NO: 9, and a VH domain comprising an amino acid sequence of SEQ I D NO: 8. In another embodiment, the binding protein comprises a constant heavy region Ig having an amino acid sequence selected from the group consisting of: SEQ I D NO: 2, and SEQ ID NO: 3; a constant light region Ig having an amino acid sequence selected from the group consisting of: SEQ ID NO: 4, and SEQ ID NO: 5; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 8; and a variable Ig light region having an amino acid sequence of SEQ ID NO: 9. In another embodiment, the binding protein comprises a constant heavy region Ig having an amino acid sequence of SEQ ID NO: 3; a constant light region Ig having an amino acid sequence of SEQ I D NO: 4; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 8, and a light variable region Ig having an amino acid sequence of SEQ ID NO: 9.
DETAILED DESCRIPTION OF THE INVENTION This invention pertains to IL-18 binding proteins, particularly anti-IL-18 antibodies, or antigen binding portions thereof, which bind thereto. Several aspects of the invention relate to antibodies and antibody fragments, and pharmaceutical compositions thereof, as well as nucleic acids, recombinant expression vectors and host cells for making such antibodies and fragments. The methods for using the antibodies of the invention to detect human IL-18, inhibit the activity of human I L-8, either in vitro or in vivo, and regulate gene expression, are also understood by the invention. This invention also pertains to a truncated IL-1 8. In related aspects, the invention also pertains to nucleic acids, recombinant expression vectors and host cells to make truncated L-1 8. Unless otherwise indicated herein, the scientific and technical terms used in connection with the present invention shall have the meanings commonly understood by those skilled in the art. In addition, unless required by the context, singular terms should include pluralities and plural terms should include singular terms. In this application, the use of "or" means "and / or" unless otherwise stated. In addition, the use of the term "including", as well as other forms, such as "includes" and "included", is not limiting. Also, terms such as "element" or "component" comprise both elements and components that comprise a unit and elements and components that comprise more than one subunit unless specifically stated otherwise. Generally, the nomenclatures used in connection with, and techniques of, tissue and cell culture, molecular biology, immunology, microbiology, genetics and nucleic acid protein and chemistry and hybridization described herein are those well known and commonly used in the art. . The methods and techniques of the present invention are generally made according to conventional methods well known in the art as described in several general and more specific references which are cited and discussed throughout the present specification unless Indicate otherwise. The purification techniques and enzymatic reactions are performed according to the manufacturer's specifications, as commonly achieved in the art or as described herein. The nomenclatures used in connection with, and the laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and pharmaceutical and medicinal chemistry described herein, are those well known and commonly used in the art. Standard techniques are used for chemical synthesis, chemical analysis, pharmaceutical preparation, formulation, and supply, and patient treatment. In order that the present invention can be understood more easily, the selected terms are defined below. The term "polypeptide" as used herein, refers to any polymeric chain of amino acids. The terms "peptide" and "protein" are used interchangeably with the term polypeptide and also refer to a polymeric chain of amino acids. The term "polypeptide" comprises artificial or natural proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide can be monomeric or polymeric. The term "isolated protein" or "isolated polypeptide" is a protein or polypeptide that by virtue of its origin or source of derivation is not associated with naturally associated components that accompany it in its natural state; it is substantially free of other proteins of the same species; it is expressed by a cell of a different species; or does not appear natural. In this way, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be "isolated" from its naturally associated components. A protein can also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art. The term "recover" as used herein, refers to the process of converting a chemical species such as a polypeptide substantially free of naturally associated components by isolation, for example, using protein purification techniques well known in the art. The term "IL-1 8", as used herein, refers to a cytokine also known as an interferon gamma inducing factor (IGIF), ie, a proinflammatory cytokine, which exhibits several functions in addition to an ability to induce gamma interferon. The term "human IL-1" used interchangeably with the term "hIL-1 8" comprises polypeptide of SEQ ID NO: 1 and fragments thereof, including but not limited to, pro-human IL-18, mature IL-18. human, and any truncated human IL-18 that retains a biological activity of IL-18 as described herein. The term "pro-human IL-18" as used herein, refers to a polypeptide of SEQ I D NO: 1. The term "human mature L-18" as used herein, refers to residues 37-193 of SEQ ID NO: 1 and the term "human truncated IL-18" as used herein, refers to residues 59-193 of SEQ ID NO: 1. Preferably, I L-1 8, and fragments thereof, are biologically active. The term "recombinant human IL-18" or "rhIL-18" as used herein, refers to human I L-1 8 generated in vitro using recombinant DNA techniques. "Biological activity of I L-1 8" as used herein, refers to all the inherent biological properties of the cytokine IL-1 8. The biological properties of 1L-1 8 include but are not limited to the binding of IL-18 receiver; promotion of maturation and activation of Th1 and Tc1 cells; promotion of the production of cytokines such as TNF, IFN? and IL-1β by various cell types; promotion of macrophages to release cytokines such as TNF and IFN ?, produce NO; promotion of FasL expression, cytotoxicity and release of cytokine (IFN?) from NK cells; cytokine promotion / cytokine release, respiratory drive, granule release, expression of adhesion molecule in neutrophils; promotion of endothelial cells to migrate and thereby promote angiogenesis; promotion of the release of GAG, MPP and production of NO in Condrocitos; promotion of COX2 expression in some cells; and reduction of cell proliferation in some cells. The terms "specific binding" or "specifically binding", as used herein, in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, mean that the interaction depends on the presence of a particular structure (for example, an antigenic determinant or epitope) in the chemical species; for example, an antibody recognizes and binds to a specific protein structure in place of generally proteins. If an antibody is specific for epitope "A", the presence of a molecule containing Epitope A (or free, unlabelled A), in a labeled "A" containing reaction and antibody, will reduce the amount of labeled A bound to the antibody . The term "antibody", as used herein, refers broadly to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional, mutant fragment , variant, or derivation thereof, which retains the binding characteristics of the essential epitope of an Ig molecule. Such derived, mutant, variant antibody formats are known in the art. The non-limiting modalities of which are discussed below. In a full-length antibody, each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three CH 1, CH 2 and CH 3 domains. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of a domain, CL. The VH and VL regions can further be subdivided into regions of hypervariability, termed complementarity determination regions (CDR), separated with regions that are more conserved structure regions called (FR). Each VH and VL is composed of three CDRs and four FRs, placed from terminal amino to terminal carboxy in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The term "antigen binding portion" of an antibody (or simply "binding portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen ( for example, hIL-18). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody modalities may also be bi-specific, specific dual, or multi-specific formats; especially binding to two or more different antigens. Examples of binding fragments comprised within the term "antigen binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH 1 domains; (ii) an F (ab ') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge in the hindered region; (iii) an Fd fragment consisting of the VH and CH 1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al. (1989) Nature 341: 544-546), which comprises a domain unique variable; and (vi) an isolated complementarity determining region (CDR). In addition, although the two domains of the Fv, VL and VH fragment are encoded for the separated genes, they can be linked, using recombinant methods, by a synthetic linker that allows them to be elaborated as a single protein chain in which the regions VL and VH are paired to form the monovalent molecules (known as single-chain Fv (scFv), see, for example, Bird et al (1988) Science 242: 423-426, and Huston et al. (1988) Proc. Nati, Acad. Sci. USA 85: 5879-5883). Such chain antibodies are also intended to be understood within the term "antigen binding portion" of an antibody. Other forms of single chain antibodies, such as diabodies are also understood. The diabodies are bivalent, bi-specific antibodies in which the VH and VL domains are expressed in a single polypeptide chain, but using a linker that is too short for equilibrium between the two domains of the same chain, thereby forcing the domains to be balanced with the complementarity domains of another chain and creating two antigen binding sites (see, for example, Holliger, P., ef al. (1 993) Proc. Nati. Acad. Sci. USA 90: 6444 -6448; Poljak, RJ, ef al. (1994) Structure 2: 1 121-1 123). Such antibody binding sites are known from the material and (Kontermann and Dubel eds., Antibodv Engineering (2001) Springer-Verlag, New York, 790 pp. (ISBN 3-540-41354-5). or part of antigen binding thereof may be part of larger immunoadhesion molecules, formed by covalent or non-covalent association of the antibody or part of the antibody with one or more proteins or peptides Examples of such immunoadhesion molecules include the use of core region of streptavidin to make a tetrameric scFv molecule (Kipriyanov, SM, ef al. (1995) Human Antibodies and Hybridomas 6: 93-1 01) and the use of a cysteine residue, a marker peptide and a polyhistidine tag terminal C to make biotinylated and bivalent scFv molecules (Kipriyanov, SM, ef al. (1994) Mol.Immunol., 311047-1 058) Antibody portions, such as Fab and F (ab ') 2 fragments, can be prepared from complete antibodies ut Using conventional techniques, such as digestion of papain or pepsin, respectively, of complete antibodies. In addition, antibodies, antibody parts and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein. An "isolated antibody", as used herein, is meant to refer to an antibody that is substantially free of other antibodies that have different antigenic specificities (eg, an isolated antibody that specifically binds L1-L8 is found substantially free of antibodies that bind specifically to antigens other than hl L-1 8). An isolated antibody that binds specifically hl L-1 8, may, however, have cross-reactivity to other antigens, such as IL-18 molecules from other species. In addition, an isolated antibody can be substantially free of cellular material and / or chemicals.
The term "human antibody", as used herein, is intended to include antibodies that have constant and variable regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by germline immunoglobulin sequences (eg, mutations introduced by site-specific or random mutagenesis in vitro or by somatic mutation in vivo), for example, in CDRs and in particular CDR2. However, the term "human antibody", as used herein, is intended to not include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, has been grafted onto human structure sequences. The term "recombinant human antibody", as used herein, is intended to include all mammalian antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a cell host (described further in Section II C, below), antibodies isolated from a recombinant human combinatorial library (Hoogenboom HR, (1 997) TIB Tech. 1 5: 62-70; Azzazy H., and Highsmith. E., (2002) Clin Biochem 35: 425-445, Gavilondo JV and Larrick JW (2002) BioTechniques 29: 128-145, Hoogenboom H., and Chames P. Immunology Today 21: 378), antibodies isolated from an animal (for example, a mouse) that is transgenic for genes of human immunoglobulin (see, for example, Taylor, LD, et al., (1992) Nuci Acids Res. 20:; SA., and Green LL (2002) Current Opinion in Biotechnology 13: 593-597; Little M. et al (2000) Immunology today 21: 364-370) or antibodies prepared, expressed, created or isolated by any other means including the division of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for Ig sequences is used, somatic mutagenesis in vivo) and thus the amino acid sequences of the VH and VL regions of recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the germline repertoire of human antibody in vivo. The term "chimeric antibody" refers to antibodies comprising light and heavy chain variable region sequences of one species and constant region sequences of another species, such antibodies having murine heavy and light chain variable regions linked to human constant regions. The term "CDR grafted anti-ruther" refers to antibodies that comprise light and heavy chain variable region sequences of a species, but in which the sequences of one or more of the CDR regions of Vh and / or VL are replaced with sequences of CDR from another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) have been replaced with CDR sequences. The term "humanized antibody" refers to antibodies comprising light and heavy chain variable region sequences of a non-human species (e.g., a mouse) but in which at least a portion of the V H and / or V L sequence is has altered to be more of "human type", that is, more similar to the variable sequences of human germ line. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding non-human CDR sequences. As used in this, the term "neutralizing binding protein hl L-1 8" refers to a protein that specifically binds hIL-18 and neutralizes a biological activity of hIL-18. Preferably, a neutralizing binding protein is a neutralizing antibody whose binding to hIL-18 results in the inhibition of a biological activity of hIL-1. Preferably, the neutralizing binding protein binds hIL-8 and reduces a biological activity of IL-1 8 by at least 20%, 40%, 60%, 80%, 85% or more. This inhibition of a biological activity of hIL-18 by a neutralizing binding protein can be assessed by measuring one or more indicators of biological activity of hIL-1 8. These indicators of biological activity of hIL-8 can be assessed by one or more of several in vitro or in vivo tests known standards in the field. The term "epitope" includes any determinant polypeptide capable of specifically binding to an immunoglobulin or T cell receptor. In certain embodiments, the determinant epitopes include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl , and in certain modalities, may have three dimensional structural characteristics, and / or specific load characteristics. An epitope is a region of an antigen that is bound by an antibody. In certain embodiments, an antibody is said to bind specifically to an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and / or macromolecules. The term "surface plasmon resonance", as used herein, refers to an optical phenomenon that is allowed for the analysis of real-time biospecific interactions by detecting alterations in protein concentrations within a biosensor binder, by example, using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51_: 19-26; Jonsson, U., et al. (1 991) Biotechniques 1 1: 620-627; Johnsson, B., ef al. (1995) J. Mol Recognit. 8: 125-131; and Johnnson, B., et al. (1991) Anal Biochem. 1 98: 268-277.
The term "Kactivo", as used herein, is intended to refer to the active scale constant for association of an antibody to the antigen to form the antibody / antigen complex as is known in the art. The term "-K¡nact¡vo", as used herein, is intended to refer to the inactive scale constant for the dissociation of an antibody from the antibody / antigen complex as is known in the art. The term "Kd", as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction as is known in the art. The term "labeled binding protein", as used herein, refers to a protein with a labeled tag that is provided for the identification of the binding protein. Preferably, the label is a detectable marker, for example, incorporation of a labeled amino acid or binding to a polypeptide of biotinyl moieties that can be detected by labeled avidin (e.g., streptavidin containing a fluorescent label or enzyme activity that can be detected by methods optical or colorimetric). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides, for example, 3 H, 14 C, 35 S, 90 Y, 99 Tc, 1 1 1 ln, 125 l, 131 l, 177 Lu, 166 Ho, or 153 Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide matches), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; polypeptide epitopes predetermined by a secondary reporter (e.g., biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine pair sequences, secondary antibody binding sites, metal binding domains, epitope tags) and magnetic agents, such as gadolinium chelates.The term "conjugated binding protein" refers to a binding protein chemically bound to a second chemical portion, such as a cytotoxic or therapeutic agent.The term "agent" as used herein denotes a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an elaborated extract of biological materials.Preferably, cytotoxic or therapeutic agents include, but do not self-mimic, pertussis toxin, taxol, cytochalasin B.gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colquicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mitramycin, actinomycin D, 1-dehydrotestosterone, glucorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. The term "crystallized binding protein" as used herein, refers to a polypeptide that exists in the form of a crystal. The crystals are of a solid state form of matter, which is different from other forms such as amorphous solid state or liquid crystalline state. Crystals are composed of groups of atoms, ions, regular, repetitive, three-dimensional molecules (eg, proteins such as antibodies), or molecular groups (eg, antigen / antibody complexes). These three-dimensional groups are placed according to the specific mathematical relationships that are well understood in the subject. The fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit. The repetition of the asymmetric unit in placement that conforms to a well-defined crystallographic symmetry, given, provide the "unit cell" of the crystal. The repetition of the unit cell by regulatory translations in all three dimensions provides the crystal. See, Giege, R and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a Practical Approach, 2nd ea.; pp. 20-1-1 6, Oxford University Press, New York, New York, (1999). "The term" polynucleotide "as used herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxynucleotides or a modified from any type of nucleotide The term includes double or single strand forms of DNA but preferably of double strand DNA The term "isolated polynucleotide" as used herein, shall mean a polynucleotide (eg, of genomic origin). , cDNA, or synthetic, or a certain combination thereof) which, by virtue of its origin, the "isolated polynucleotide" is not associated with all or a part of a polynucleotide with which the "isolated polynucleotide" is found in nature; it is operably linked to a polynucleotide that does not bind to the in-nature, and does not occur in nature as part of a larger sequence.The term "vector", as used herein, is so that it refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been bound. One type of vector is a "plasmid," which refers to a circular double-stranded DNA cycle in which additional DNA segments can be ligated. Another type of vector is a viral vector, where additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous duplication in a host cell into which they are introduced (eg, bacterial vectors having duplication bacterial origin and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby duplicated together with the host genome. In addition, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, "expression vectors." In general, expression vectors useful in recombinant DNA techniques are often found in the form of plasmids. specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector, however, it is intended that the invention includes such other forms of expression vectors, such as viral vectors (e.g., retroviruses defective duplications, adenoviruses and adeno-associated viruses), which serve equivalent functions The term "operatively linked" refers to a juxtaposition wherein the components described are in a relationship that allows them to function in their intended manner. control "operably linked" to a coding sequence is ligated in such a way that the expression of the sequence dificadora is achieved under conditions compatible with the control sequences. "Operably linked" sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term "expression control sequence" as used herein refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are linked. Expression control sequences include initiation of proper transcription, termination, enhancer and promoter sequences; efficient RNA processing signals such as division and polyadenylation signals; sequences that house the cytoplasmic mRNA; sequences that improve translation efficiency (ie, Kozak consensus sequence); sequences that improve protein stability; and when desired, sequences that improve protein secretion. The nature of such control sequences differs depending on the host organism; in prokaryotes, such control sequences generally include ribosomal binding site, promoter, and translation termination sequence; in eukaryotes; generally, such control sequences include promoters and transcription termination sequence. The term "control sequences" is intended to include components whose presence is essential for expression and processing, and may also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. "Transformation", as defined herein, refers to any process by which exogenous DNA enters a host cell. The transformation can occur under artificial or natural conditions using various methods well known in the art. The transformation may depend on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic and eukaryotic host cell. The method is selected on the basis of the host cell that is transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such "transformed" cells include stably transformed cells in which the inserted DNA is capable of duplication either as a self-replicating plasmid or as part of the host chromosome.
They also include cells that transiently express the inserted DNA or RNA for limited periods of time. The term "recombinant host cell" (or simply "host cell"), as used herein, is meant to refer to a cell in which the exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell, but to the offspring of such a cell. Because certain modifications can occur in successor generations due to either mutation or environmental influences, such offspring may not, in fact, be identical to the main cell, but are still included within the scope of the term "host cell" as used herein. Preferably, the host cells include eukaryotic or prokaryotic cells selected from any of the life realms. Preferred eukaryotic cells include, animal, plant, fungal and protist cells. More preferably, host cells include but are not limited to prokaryotic cell strain E. coli; mammalian cell strains CHO and COS; the Sf9 insect cell line; and the fungal cell Saccharomyces cerevisae. Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques can be performed according to the manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and methods can generally be performed according to conventional methods well known in the art and as described in several general and more specific references that are cited and discussed throughout the present specification. See, for example, Sambrook ef al. , Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y. (1989)), which is incorporated herein by reference for any purpose. "Transgenic organism", as known in the art and as used herein, refers to an organism having cells that contain a transgene, wherein the transgene introduced into the organism (or an ancestor of the organism) expresses a polypeptide not naturally expressed in the organism. A "transgene" is a DNA construct, which is stable and operatively integrated into the genome of a cell from which a transgenic organism develops, directing the expression of the gene product encoded in one or more cell types or tissues of the transgenic organism. The term "regular" and "modular" are used interchangeably, and, as used herein, refers to a change or alteration in the activity of a molecule of interest (e.g., the biological activity of hIL-18) . The modulation may be an increase or a reduction in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction. Correspondingly, the term "modulator", as used herein, is a compound capable of changing or altering an activity or function of a molecule of interest (e.g., the biological activity of hIL-18). For example, a modulator can cause an increase or reduction in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator. In certain embodiments, a modulator is an inhibitor, which reduces the magnitude of at least one activity or function of a molecule. Exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described, for example, in WO01 / 83525. The term "combatant", as used herein, refers to a modulator which, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the combatant. Particular combatants of interest may include, but are not limited to, I L-18 polypeptides or polypeptides, nucleic acids, carbohydrates, or any of the other molecules that bind to hl L-1. 8. The term "antagonist" or " "inhibitor", as used herein, refers to a modulator which, when contacted with a molecule of interest causes a reduction in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist. The particular antagonist of interest includes those that block or modulate the biological or immunological activity of hl L-18. Antagonists or inhibitors of hIL-18 may include, but are not limited to, proteins, nucleic acids, carbohydrates, or any of the other molecules that bind to I L-18. The term "sample", as used herein, is used in the broadest sense. A "biological sample", as used herein, includes, but is not limited to, any amount of a substance of a living thing or formally living thing. Such living things include, but are not limited to, humans, mice, rats, monkeys, dogs, rabbits and other animals. Such substances include, but are not limited to, blood, serum, urine, synovial fluid, cells, organs, tissues, bone marrow, lymph nodes and spleen. The term "competes" as used herein, and as it is generally known and used by those skilled in the art, refers to the ability of a binding protein to interfere with, or otherwise hinder the binding of a second binding protein to a common binder for both binding proteins (e.g., I L-1 8). Useful assays for determining the competition characteristics of the binding proteins are well known in the art. Preferred competition tests are described herein.
I. Human Antibodies that Link Human IL-18. One aspect of the present invention provides isolated human antibodies, or antigen binding portions thereof, which bind to IL-18 with high affinity, a low inactive scale and high neutralizing capacity. Preferably, the antibodies, or parts thereof, are isolated antibodies. Preferably, the human antibodies of the invention are neutralizing anti-human IL-18 antibodies.
A. Method for Making Anti-IL-18 Antibodies The antibodies of the present invention can be made from any of a number of techniques known in the art. A particularly preferred method for generating anti-IL-18 antibodies of the invention includes using XENOMOUSE transgenic mice, and using hybridoma and SLAM cellular manipulation techniques (Abgenix, Inc., Fremont, CA) known in the art to prepare antibodies, and use antigens comprising peptide I L-1 8 described in Example 3.2, ie, amino acid sequence comprising human IL-1 8 of SEQ ID NO. 1 and fragments thereof. In one embodiment of the present invention, antibodies are produced by immunizing a non-human animal comprising some or all of the immunoglobulin site with an IL-1 antigen 8. In a preferred embodiment, the non-human animal is a mouse transgenic XENOMOUSE, an elaborate mouse strain comprising large fragments of the human immunoglobulin site and is deficient in the production of mouse antibody. See, for example, Green et al. , Nature Genetics 7: 1 3-21 (1994) and U.S. Patents 5,916,771, 5,939,598, 5,985.61 5, 5,998,209, 6,075, 1 81, 6,091, 001, 6, 1 14,598 and 6, 30,364. See also WO 91/10741, published July 25, 1991 WO 94/02602, published February 3, 1994, WO 96/34096 and WO 96/33735, both published October 31, 1 996, WO 98 / 16654 published April 23, 1998, WO 98/24893, published June 1, 1998, WO 98/50433, published November 12, 1998, WO 99/45031, published September 10, 1999 , WO 99/53049, published on October 21, 1999, WO 00 09560, published on February 24, 2000 and WO 00/037504, published on June 29, 2000. The transgenic mouse XENOMOUSE produces an adult type human repertoire of fully human antibodies, and generates human antigen-specific Mabs. The XENOMOUSE transgenic mouse contains approximately 80% of the human antibody repertoire through the introduction of dimensioned megabase, YAC fragments of germline configuration of the human heavy chain site and x light chain site. See, Méndez ef al. , Nature Genetics 15: 146-156 (1997), Green and Jakobovits J. Exp. Med. 188: 483-495 (1998), the descriptions of which are incorporated in such a way for reference. The invention also provides a method for making anti-I L-18 antibodies from non-human, non-mouse animals by immunizing non-human transgenic animals that comprise the human immunoglobulin site. One can produce such animals using the methods immediately described above. The methods described in these patents can be modified as described in U.S. Patent 5,994,619. In a preferred embodiment, the non-human animals may be rats, sheep, pigs, goats, cattle or horses. In another embodiment, the non-human animal comprising the human immunoglobulin gene site are animals that have a "minisite" of human immunoglobulins. In the minisite procedure, an exogenous Ig site is mimicked through the inclusion of the individual Ig site genes. In this manner, one or more VH genes, one or more DH genes, one or more JH genes, a mu constant region, and a second constant region (preferably, a gamma constant region), are formed in a construct for insertion into a mammal. This procedure is described, inter alia, in U.S. Pat. UU No. 5, 545,807, 5, 545,806, 5,625,825, 5,625, 126, 5, 633,425, 5,661, 016, 5,770,429, 5,789,650, 5,814,318, 5,591, 669, 5,612,205, 5,721, 367, 5,789.21 5, and 5,643,763, incorporated in such a way to reference. One advantage of the minisite method is the rapidity with which constructs that include parts of the Ig site can be generated and introduced into animals. However, a potential disadvantage of the minisite method is that there may not be enough diverse immunoglobulin to support the development of complete B cell, so that there may be low antibody production. In order to produce a human anti-IL-1 antibody, a non-human animal comprising some or all of the human immunoglobulin site is immunized with an IL-1 antigen 8 and the antibody or the antibody producing cell is isolated from the animal. . The antigen I L-1 8 can be I L-1 8 isolated and / or purified and is preferably a human I L-1 8. In another embodiment, the IL-1 antigen 8 is a fragment of I L-1 8, preferably mature IL-18. In another embodiment, the IL-18 antigen is a fragment comprising at least one epitope of IL-18. The immunization of animals can be done by any method known in the art. See, for example, Harlow and Lane, Antibodies: Laboratory Manual, New York: Cold Spring Harbor Press, 1 990. Methods for immunizing non-human animáis such as mice, rats, sheep, goats, pigs, cattle and horses are known in the material. See, for example, Harlow and Lane and U.S. Patent 5,994,619. In a preferred embodiment, the I L-18 antigen is administered with an adjuvant to stimulate the immune response. Such adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulatory complexes). Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local reservoir, or may contain substances that stimulate the host to secrete factors that are chemotactic for macrophages and other components of the immune system. Preferably, if a polypeptide is being administered, the immunization schedule will include two or more administrations of the polypeptide, spreading for weeks. The example 2.2. A provides a protocol for immunizing a XENOMOUSE transgenic mouse with human I L-1 8 in phosphate-regulated saline.
B. Production of Antibodies and Cell Strains Antibody Producing After Immunization of an Animal with an Antigen I L-18, the antibodies and / or antibody-producing cells can be obtained from the animal. A serum containing anti-I L1 8 antibody is obtained from the animal by bleeding or slaughter of the animal. The serum can be used as if it were obtained from the animal, and the immunoglobulin fraction can be obtained from the serum, or the anti-I L-18 antibodies can be purified from the serum. The serum or immunoglobulins obtained in this way are polyclonal, thus having a heterogeneous group of properties. In another embodiment, immortalized antibody producing hybridomas can be prepared from the immunized animal. After immunization, the animal is sacrificed and spleen B cells are fused into immortalized myeloma cells as is well known in the art. See, for example, Harlow and Lane, supra. In a preferred embodiment, the myeloma cells do not secrete immunoglobulin polypeptides (a non-secretory cell line).
After the fusion and antibiotic selection, the hybridomas are selected using IL-18, or a part thereof, or a cell expressing IL-18. In a preferred embodiment, the initial screening is performed using enzyme-linked immunoassay (ELISA) or a radioimmunoassay (RIA), preferably an ELISA. An exemplary ELISA selection is provided in WO 00/37504, incorporated herein by reference. Hybridomas producing anti-IL-18 antibody are selected, cloned and further chosen for desirable characteristics, including robust hybridoma growth, high antibody production and desirable antibody characteristics, as discussed below. Hybridomas can be cultured and expanded in vivo in syngeneic animals, in animals lacking an immune system, for example, nude mice, or in cell culture in vivo. The methods for selection, cloning and expansion of the hybridomas are well known to those skilled in the art. Preferably, the immunized animal is a non-human animal expressing human immunoglobulin genes and the spleen B cells are fused to a myeloma derived from the same species as the non-human animal. More preferably, the immunized animal is a XENOMOUSE transgenic mouse and the myeloma cell lineage is a non-secreting mouse myeloma, such as the myeloma cell line is P3X63Ag8.653 (see, for example, Example 2.2.B). In one aspect, the invention provides hybridomas that produce human anti-L1-8 antibodies. In a preferred embodiment, the hybridomas are mouse hybridomas, as described above. In another preferred embodiment, the hybridomas are produced in a non-human, non-mouse spice such as rats, sheep, pigs, goats, cattle or horses. In another embodiment, hybridomas are human hybridomas, in which a non-secretory human myeloma is fused with a human cell expressing an anti-IL-18 antibody. In another aspect of the invention, recombinant antibodies are generated from single, isolated lymphocytes, using a method referred to in the art as the selected lymphocyte antibody (SLAM) method, as described in US Pat. UU DO NOT. 5,627, 052, PCT Publication WO 92/02551 and Babcock, J. S. et al. (1 996) Proc. Nati Acad. Sci. USA 93: 7843-7848. In this method, single cells that secrete antibodies of interest, for example, lymphocytes derived from any of the immunized animals described in Section 1 (A), are selected using an antigen-specific hemolytic plaque assay, wherein the 1L antigen -18, or a fragment thereof, is coupled sheep red blood cells using a linker, such as biotin, and used to identify the unique cells that secrete antibodies with specificity for I L-18. Next to the identification of antibody secreting cells of interest, light and heavy chain variable region cDNAs are rescued from cells by reverse transcriptase-PCR and these variable regions can be expressed, in the context of suitable immunoglobulin constant regions, (e.g., human constant regions), in host cells mammal, such as COS or CHO cells. The cells transferred with the amplified immunoglobulin sequences, derived from lymphocytes selected in vivo, can thus overcome the additional analysis and in vitro selection, for example, by turning the transfected cells to isolate the cells expressing antibodies to IL-18. Amplified immunoglobulin sequences can be manipulated in vitro, such as by in vitro affinity maturing methods such as those described in PCT Application WO 97/291 31 and PCT Publication WO 00/56772. The in vitro methods can also be used to make the antibodies of the invention, wherein an antibody library is selected to identify an antibody having the '15 desired binding specificity. Methods for such selection of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al. , U.S. Pat. UU No. 5,223,409; Kang ef al. PCT Publication No. WO 92/1861 9;; Dower et al. PCT Publication No. WO 91/17271; Winter ef al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/1 5679; Breitling ef al. PCT Publication No. WO 93/01288; McCafferty ef al. PCT Publication No. WO 92/01 047; Garrard ef al. PCT Publication No. WO 92/09690; Fuchs ef al. (1 991) Bio / Tecnology 9: 1370-1 372; Hay et al. (1 992) Hum Antibod Hybridomas 3: 81 -85; Huse ef al. (1 989) Science 246: McCafferty ef al. Nature (1990) 348: 552-554; Griffiths I went to. (1993) EMBO J 12: 725-734; Hawkins et al. J Mol Biol (1992) 226: 889-896; Clackson ef al. (1991) Nature 352: 624-628; I recorded al. (1992) PNAS 89: 3576-3580; Garrad e to al. (1991) Bio / Technology 9: 1 373-1377; Hoogenboom ef al. (1991) Nuc Acid Res 1_9: 4133-41 37; and Barbas ef al. (1 991) P? / \ S 88: 7978-7982, US patent application publication. U.S. 200301 86374, and PCT Publication No. WO 97/29131, the contents of which are incorporated herein by reference. The recombinant antibody library can be from a subject immunized with I L-1 8, or a part of I L-1 8. Alternatively, the recombinant antibody library can be from a natural subject, i.e., one that has not been immunized with I L-1 8, such as a library of human antibody from a human subject that has not been immunized with human I L-18. The antibodies of the invention are selected by selecting the recombinant antibody library with the peptide comprising human IL-18 (e.g., a peptide corresponding to a part of hl L-18) to select such antibodies that recognize IL-18. . The methods for conducting such selection and choice are well known in the art, as described in the references in the preceding paragraph. To select the antibodies of the invention which have binding affinities in particular for IL-18, such as those dissociated from human I L-1 8 with a particular klnactivity scale constant, the method known in the art of resonance of Surface plasmon can be used to select the antibodies having the K-scale constant, Nactiva- To select the antibodies of the invention having a particular neutralizing activity for hIL-18, such as those with a particular IC50, the well-known standard methods In the matter to assess the inhibition of hIL-18 activity can be used. In one aspect, the invention pertains to an isolated antibody, or an antigen binding portion thereof, that binds human IL-18. Preferably, the antibody is a neutralizing antibody. Preferably, the antibody is a human antibody. In various embodiments, the antibody is a recombinant antibody or a monoclonal antibody. The most preferred neutralizing antibody of the invention is referred to herein as 2.5 (E) and has VL with amino acid sequence of SEQ I D NO: 7 and VH with amino acid sequence of SEQ I D NO: 6. More preferably, antibody 2.5 (E) binds human I L-18 with a K of less than 5x1 010M (see Example 2.2 F). Preferably, the anti-IL-18 antibodies of the present invention, such as antibody 2.5 (E) and related antibodies, show an ability to reduce or neutralize the activity of IL-1 8, for example, as assessed by either of various in vitro or in vivo assays known in the art (for example, see Example 3.2, F). For example, these antibodies neutralize the IL-18-induced production of human gamma interferon in KG-1 cells with IC50 values in the range of at least about 10 0 8M, about 10 9M, or about 10 10M. these antibodies also neutralize the production induced by I L-1 8 of gamma human interferon in total blood cells with IC0 values in the range of at least about 1 0"8M, approximately 1 0"9M, or approximately 10" 10M. In a particularly preferred embodiment, anti-IL-18 antibody 2.5 (E) binds to human IL-18 in various forms, including pro-IL-1 8, mature L-1 8 and truncated IL-18. Antibody 2.5 (E) does not bind specifically to other cytokines, such as I L-2, I L-3, IL-4, I L-5, IL-6, IL-7, I L-8, I L -9, I L-1 0, IL-1 1, IL-12, I L-13, I L-15, I L-16, IL-17, IL-21, TNF, LT (lymphotoxin), LT 1 ß2, and LTa2ßl However, antibody 2.5 (E) shows cross-reactivity for IL-1 8 of another species. For example, the antibody neutralizes the activity of 1L-18 cynomologo monkey (IC50 for clumping I L-1 8 = 9.1 E X 10"1 1; See example 2.2.J1). In one aspect, the invention pertains to antibodies 2.5 (E) and functional antibody portions, and other human antibodies and functional antibody portions with equivalent properties for 2.5 (E), such as high affinity binding for I L-1 8 with low dissociation kinetics and high neutralizing capacity. In preferred embodiments, the isolated antibody, or antigen-binding portion thereof, binds human IL-18, wherein the antibody, or antigen-binding portion thereof, dissociates from human IL-18 at a scale constant kActiVa of about 0.1 s "1 or less, as determined by surface plasmon resonance, or which inhibits human IL-1 8 activity with an IC50 of about 1 x 10" 6M or less. Alternatively, the antibody, Or an antigen binding portion thereof, can be dissociated from human IL-1 8 with a Kinactava constant of about 1 x 10"2s" 1 or less, as determined by surface plasmon resonance, or can inhibit the activity of human IL-18 with a 1C50 of about 1 x 10"7M or less.Alternatively, the antibody, or an antigen-binding portion thereof, can be dissociated from human I L-1 8 with a kinetic-scale constant of about 1 or 1 0"3s" 1 or less, as determined by the plasmon resonance of surface, or can inhibit the activity of human IL-18 with an IC50 of about 1 x 10"7M or less. Alternatively, the antibody, or an antigen-binding portion thereof, can be dissociated from human I L-18 with a kinetic-scale constant of about 1 x 1 0"4 s" 1 or less, as determined by surface plasmon resonance, or can inhibit the activity of human IL-18 with an IC50 of about 1 x 10"9M or less.Alternatively, the antibody, or an antigen-binding portion thereof, can be dissociated from I L -1 8 human with a scale constant k na ct ct _ V a a de a of approximately 1 x 1 0"5 s" 1 or less, as determined by surface plasmon resonance, or can inhibit the activity of human IL-18 with an IC 50 of about 1 x 1 0"10M or less. Alternatively, the antibody, or an antigen-binding portion thereof, can be dissociated from human I L-1 8 at a steady-state constant of about 1 x 10"5s" 1 or less, as determined by resonance. surface plasmon, or can inhibit the activity of human IL-18 with an IC 50 of about 1 x 10"11M or less.Although in another embodiment, the invention provides an isolated human antibody, or an antigen binding portion thereof. , with a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO : 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 33; SEQ ID NO: 35; SEQ ID NO: 37; SEQ ID NO: 39; or SEQ ID NO: 41; and a variable region of heavy chain (VH) comprising an amino acid sequence SEQ ID NO: 6; NO: 8; SEQ ID NO: 10; SEQ ID NO: 12; SEQ ID NO: 14; SEQ ID NO : 16; SEQ ID NO: 18; SEQ ID NO.20; SEQ ID NO: 22; SEQ ID NO: 24; SEQ ID NO: 26; SEQ ID NO: 28; SEQ ID NO: 30; SEQ ID NO: 32; SEQ ID NO: 34; SEQ ID NO: 36; SEQ ID NO: 38 or SEQ ID NO: 40. In certain embodiments, the antibody comprises a constant region lgG1, IgG3, IgG3, IgG4, IgA, IgE, IgM or IgD. Preferably, the heavy chain constant region is a lgG1 heavy chain constant region or a lgG4 heavy chain constant region. In addition, the antibody may comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region.
Preferably, the antibody comprises a kappa light chain constant region. Alternatively, the antibody part can be, for example, a Fab fragment or a single chain Fv fragment. Replacements of amino acid residues in the Fe part to alter antibody effector function are known in the art (Winter, et al., U.S. Patent No. 5,648,260, 5624821). The Fe part of an antibody mediates several important effector functions, eg, cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life / scale of antibody clearance and antigen-antibody complexes. In some cases, these effector functions are desirable for the therapeutic antibody but in other cases it may be necessary or even harmful, depending on the therapeutic objectives. Certain isotypes of human IgG, particularly lgG 1 and lgG3, mediate ADCC and CDC by binding to Fc? Rs and Clq complement, respectively. Neonatal Fe receptors (FcRn) are the critical components that determine the circulating half-life of antibodies. In yet another embodiment at least one amino acid residue is replaced in the constant region of the antibody, for example, the Fe region of the antibody, such that the effector functions of the antibody are altered. One embodiment provides a labeled binding protein wherein one antibody or part of the antibody of the invention is derived from or linked to another functional molecule (e.g., another peptide or protein). For example, a labeled binding protein of the invention can be derived by functionally binding an antibody or part of the antibody of the invention (by chemical coupling, genetic fusion, non-covalent association or otherwise) to one or more other molecular entities, such as another antibody (for example, a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and / or a protein or peptide that can mediate the association of the antibody or part of the antibody with another molecule (such as as a core region of streptavidin or a polyhistidine tag). Useful detectable agents with which an antibody or part of the antibody of the invention can be derived, include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-naphthalenesulfonyl chloride, phycoerythrin, and the like. An antibody can also be derived with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, oxidase glucose and the like. When an antibody is derived with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable horseradish peroxidase agent is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody can also be derived with biotin, and detected through the indirect measurement of avidite or streptavidin binding. Another embodiment of the invention provides a crystallized binding protein. Preferably, the invention relates to crystals of complete anti-IL-18 antibodies and fragments thereof as described herein, and formulations and compositions comprising such crystals. In one embodiment, the crystallized binding protein has a longer in vivo half-life different from the soluble counterpart of the binding protein. In another embodiment, the binding protein retains the biological activity after crystallization. The crystallized binding protein of the invention can be produced according to methods known in the art and as described in WO 02072636, incorporated herein by reference (See also Example 2.2.M). Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues. The production of nascent in vivo protein can overcome the additional processing, known as post-translational modification. In particular, sugar (glycosyl) residues can be added enzymatically, a process known as glycosylation. The resulting proteins that carry covalently bound oligosaccharide side chains are known as glycosylated proteins or glycoproteins. Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms can produce different glycosylation enzymes (eg, glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and glycosyl residue composition, may differ from the host system in which the particular protein is expressed. Preferably, the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human. It is known to those skilled in the art that different protein glycosylation can result in different protein characteristics. For example, the efficacy of a therapeutic protein produced in a host of microorganism, such as yeast, and glycosylated, using the endogenous path of yeast can be reduced compared to that of the same protein expressed in a mammalian cell, such as a cell line CHO. Such glycoproteins can also be immunogenic in humans and show reduced half-life in vivo after administration. Specific receptors in humans and other animals can recognize specific glycosyl residues and promote rapid elimination of the protein from the bloodstream. Other side effects may include changes in protein fold, solubility, susceptibility to proteases, trafficking, transport, compartment, secretion, recognition by other proteins or factors, antigenicity, or allergy. According to the foregoing, an expert can a therapeutic protein with a specific composition and glycosylation model, for example, glycosylation composition and model identical, or at least similar, to that produced in human cells or in species-specific cells of the animal the intended subject. The expression of glycosylated proteins different from that of a host cell can be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. The use of techniques known in the art by the skilled artisan can generate antibodies or antigen binding portions thereof showing glycosylation of human protein. For example, the yeast strains have been genetically modified to express glycosylation enzymes that occur not naturally such that the glycosylated proteins (glycoproteins) produced in these yeast strains show proteinic glycosylation identical to that of animal cells, especially human cells (US patent applications 2004001 8590 and 20020137134). In addition, it will be appreciated by a person skilled in the art that a protein of interest can be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that host cells member of the library produce the protein of interest with models of glycosylation variants. An expert can thus choose and isolate the protein of interest with particular novel glycosylation models.
Preferably, the protein having a particularly selected new glycosylation pattern shows altered or improved biological properties.
C. Production of recombinant IL-18 antibodies The antibodies of the present invention can be produced by any of a number of techniques known in the art. For example, expression of host cells, wherein the expression vector (s) encoding the light and heavy chains is transfected into a host cell by standard techniques. The various forms of the term "transfection" is intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into eukaryotic or prokaryotic host cells, for example, electroporation, calcium-phosphate precipitation, transfection of DEAE-dextran and Similary. Although it is possible to express the antibodies of the invention in either eukaryotic or prokaryotic host cells, the expression of antibodies in eukaryotic cells is preferable, and more preferable in mammalian host cells, because such eukaryotic cells (and in cells of particular mammal) are more likely than prokaryotic cells to group and secrete an immunologically active and properly folded antibody. Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Uriaub and Chasin, (1980) Proc. Nati. Acad. Sci. USA 77: 4216-4240, used with an eligible DHFR marker, for example, as described in RJ Kaufman and PA Sharp (1982) Mol. Biol. 159: 601-621), NSO myeloma cells, COS cells and SP2 cells. When the recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for antibody expression in the host cells or, more preferably, secretion. of the antibody in the culture medium in which the host cells grow. The antibodies can be recovered from the culture medium using standard protein purification methods. The host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations in the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding either light chain and / or heavy chain functional fragments of an antibody of this invention. Recombinant DNA technology can also be used to remove something, or all of the DNA that encodes either one or both heavy and light chains that is not necessary for binding to the antigens of interest. The expressed molecules of such truncated DNA molecules are also comprised by the antibodies of the invention. In addition, bifunctional antibodies can be produced in which one light chain and one heavy chain are an antibody of the invention and the light chain and another heavy chain are specific for a different antigen from the antigens of interest by degrading an antibody of the invention to a second antibody by standard chemical degradation methods.
Table 1 is a list of amino acid sequences of VH and VL regions of anti-hLL-1 8 preferred antibodies of the ^ Q invention. In the VH region, the amino acid that occurs naturally at position 1 of the amino terminal (N-terminal) is either Glutamate (E) or Glutamine (Q). However, to generate the recombinant protein with homogeneous N-terminal during the large-scale production of protein comprising VH region, ^ 5 Glutamate (E) is preferred at position 1 of the N terminal.
Table 1 List of amino acid sequences of the VH and VL regions 0 Protein Region Sequence identification Protein sequence 12345678901234567890 VL 2.5 CDR- Residues 24-34 of SEQ ID L1 NO ..: 7 RASESISSNLA VL 2.5 CDR- Residues 50-56 of SEQ ID L2 NO ..: 7 TASTRAT VL 2.5 CDR- Residues 89-98 of SEQ ID L3 NO. .: 7 QQYNNWPSIT QVQLQESGPGLVTPSQTLSL TCTVSGGSISSGGHYWTWIR QHPGKGLEWIGYIYYSGSTY VH 2.13 (E) SEQ ID NO ..: 8 YNPSLKSRLTISVDTSKNQF SLKLSSVAAADTAVYYARD RGGSGSYWDYWGQGTLVTVS S VH 2.13 Residues 31-37 of SEQ ID CDR-H1 NO ..: 8 SGGHYWT VH 2.13 Residues 52-67 of SEQ ID CDR-H2 NO ..: 8 YIYYSGSTYYNPSLKS VH 2.13 Residues 100-110 of SEQ CDR-H3 ID NO. .: 8 DRGGSGSYWDY EIVLTQSPGTLSLSPGERAT LSCRGSRSVSSGYLAWYQQK PGQAPRLLIYGVSIRATGIP VL 2.13 SEQ ID NO ..: 9 DRFSGSGSGTDFTLTISRLE PEDFAVYYCQQYHGSPLTFG GGTKVEIKR VL 2.13 Residues 24-35 of SEQ ID CDR-L1 NO ..: 9 RGSRSVSSGYLA VL 2.13 Residues 21-27 of SEQ ID CDR-L2 NO ..: 9 GVSIRAT VL 2.13 Residues 90-98 of SEQ D CDR-L3 NO. .: 9 QQYHGSPLT QVQLQESGPGLVKPSETLSL TCTVSGGSIRNYYWSWIRQP VH 2.3 SEQ ID NO ..: 10 PGKGLEWVGYIYSSGSTNYN PSLKSRVTISVDTSKNQFSL KLSSVTAADTAVYYCARDRG GASFFDYWGQGTLVTVSS VH 2.3 Residues 31-35 of SEQ ID CDR-H1 NO ..: 10 NYYWS VH 2.3 Residues 50-65 of SEQ ID YIYSSGSTNYNPSLKS Protein Region Identification Sequence protein sequence 12345678901234567890 EIVLTQSPGTLSLSPGERAT LSCRASRSLSSGYLAWYQQK VL 215 PGQAPRLLIYGASIRATGIP SEQ ID NO ..: 13 DRFSGSGSATDFTLTISRLE PEDFAVYYCQQYNYSPLTFG GGTRVEINR VL 215 CDR- Residues 24-35 of SEQ ID L1 NO ..: 13 RASRSLSSGYLA VL 215 CDR- Residues 51-57 of SEQ ID L2 NO ..: 13 GAS I RAT VL 215 CDR- Residues 90-98 of SEQ ID L3 NO ..: 13 QQYNYSPLT EVQLVESGGGSVQPRGSLRL SCAASGFTFSSYSMNWVRQA PGKGLEWVSYFSSSGGIIYY VH 231 SEQ ID NO ..: 14 ADSVKGRFTISRDNAKNSLY LQMNSLRDEDTAVYYCARDD SSGYYPYFFDYWGQGTLVTV SS VH 231 Residues 31-35 of SEQ ID CDR-H1 NO ..: 14 SYS N VH 231 Residues 50-66 of SEQ ID CDR-H2 NO ..: 14 YFSSSGGIIYYADSVKG VH 231 Residues 99-111 of SEQ ID CDR-H3 NO .. 4 DDSSGYYPYFFDY DIVMTOSPDSLAVSLGERAT INCKSSQTVLYRSNNKNYLA VL 231 WYQQKSGQPPKLLIYWASTR SEQ ID NO ..: 15 ESGVPDRFSGSGSGTDFTLT ISSLQAEDVAVYYCQQYYST PLTFGGGTKVEIKR VL 231 CDR- Residues 24-40 of SEQ ID L1 NO ..: 15 KSSQTVLYRSNNKNYLA VL 231 CDR- Residues 56-62 of SEQ ID L2 NO ..: 15 WASTRES VL 231 CDR- Residues 95-103 of SEQ D L3 NO ..: 15 QQYYSTPLT QLQLQESGPGLVKPSETLSL TCTVSGGSISSRVYYWGWIR VH 251 QPPGKGLEWIGSIYYSGSTY SEQ ID NO ..: 16 YNPSLKSRVTISVDASKNQF SLKLSSVTAADTAIYYCARE DSSAWVFEHWGQGTLVTVSS VH 251 Residues 31-37 of SEQ ID CDR-H1 NO ... -16 SRVYYWG VH 251 Residues 52-67 of SEQ ID CDR-H2 NO ..: 16 SIYYSGSTYYNPSLKS VH 251 Residues 100-109 of SEQ CDR-H3 ID NO ..: 16 EDSSAWVFEH EIVLTQSPDTLSLSPGERAT LSCRASHILSRNYLAWYQQK PGQAPRLLMYGISIRATGIP SEQ ID NO ..: 17 VH 215 DRFSGSGSGADFTLTINRLE PEDFAVYYCQHYDNSLCSFG QGTKLEVKR VL 251 CDR- Residues 24-35 of SEQ ID L1 NO ..: 17 RASHILSRNYLA VL 251 CDR- Residues 51-57 of SEQ ID L2 NO ..: 17 GISIRAT VL 251 CDR- Residues 90-98 of SEQ ID L3 NO. .: 17 QHYDNSLCS Protein Region Identification Protein sequence sequence 12345678901234567890 CDR-L2 NQ ..: 29 VL 444 Residues 90-98 of SEQ D CDR-L3 NO ..: 29 QQYGYSPLT QVQLQESGPGLVKPSQTLSL TCTVSGGSISSGGHYWSWIR VH 478 SEQ ID NO ..: 30 QHPGKGLEWIGYIYYSGSTH YNPSLKSRVTISVDTSKNQF SLKLRSVSAADTAGYYCASL YNGNGYFDLWGRGTLVTVSS VH 478 Residues 31-37 of SEQ ID CDR-H1 NO ..: 30 SGGHYWS VH 478 Residues 52-67 of SEQ ID CDR-H2 NO ..: 30 YIYYSGSTHYNPSLKS VH 478 Residues 99-109 of SEQ ID CDR-H3 NO. .: 30 SLYNGNGYFDL EIVLTQSPGTLSLSPGERAT LSCRASQSISSGYLAWYQQK VL 478 SEQ ID NO ..: 31 PGQAPRLIIYGVSRRATGIP DRFSGSGSGADFTLTISRLD PEDFVVYYCQQYGFSPLTFG GGTKVEIKR VL 478 Residues 24-35 of SEQ ID CDR-L1 NO ..: 31 RASQSISSGYLA VL 478 Residues 51-57 of SEQ ID CDR-L2 NO ..: 31 GVSRRAT VL 478 Residues 90-98 of SEQ ID CDR-L3 NO. .: 31 QQYGFSPLT QLQLQESGPGLVKPSETLSL TCTVSGGSISRSYDYWGWIR SEQ ID NO ..: 32 QPPGKGLEWIGSIYYRGSTY VH 521 YNPSLKSRVTISVDTSKNQF SLKLSSVTAADTAVYYCARE YSTTWSIDYWGQGTLVTVSS VH 521 Residues 31-37 of SEQ ID CDR-H1 NO ..: 32 RSYDYWG VH 521 Residues 52-67 of SEQ ID CDR-H2 NO ..: 32 SIYYRGSTYYNPSLKS VH 521 Residues 100-109 of SEQ CDR-H3 ID NO. .: 32 EYSTTWSIDY L3 ID NO ..: 37 The preceding isolated anti-IL-18 antibody CDR sequences establish a new family of I-L-18 binding proteins, isolated according to this invention, and comprising polypeptides that include the CDR sequences listed in Table 2 below. To generate and select CDR's of the invention having preferred IL-1 neutralizing and / or binding activity, standard methods known in the art for generating binding proteins of the present invention and assessing IL-18 binding and / or neutralizing the characteristics of those binding proteins can be used, including but not limited to those specifically described herein.
TABLE 2: Condensing affinity CDR consensus IL-18 (The alternative residues are listed below each amino acid position, - indicates the possible absence of residue) ReIdentifiSequence Conduction gión cado, of CDR sequence CDR- SEQ ID i? 2? 3 X. Xs? 6 X7 H 1 NO ..: 2 S Y w I G - - N G G H Y w T H R Y W S s R S D Y N G Y C S M H V L I D CDR- SEQ ID Xi? 2 Xa X. Xs? S X7 X? X. X1 X X X X X X X H2 NO ..: 3 0 1 1 F I Y P G D S E T R Y S P T F Q - Y F S Y S G T T Y Y N P S L K S G H w s R G I N s S A D S V K S D R N I V V K H CDR- SEQ ID Xi? 2? 3 X. Xs X? 7 X X X. X1 X X X X X X X X H3 NO ..: 44 1 15 1 1 S 7 8 V G s G W Y P Y T D R G S s G S F w F D I Y Y G M D V E D Y Y A S F D D D Y D S S R N G F Y P L Y F Y C K T Y W V M Y H L D T N G I E V Y W N P Y A V F G CDR- SEQ ID Xi? 2 X? X? X? X? X? X? X? 9 X1 X X X X X X X L1 NO ..: 45 1 16 1 5 7 R A s E s I S S N L A K G R I V G G G Y L A K N Y L A S Q T L L Y Y S N N T Y C D H N F N R R D V E N T Y R N S G K H D G D CDR- SEQ ID Xi? 2 X? X? X? X? X7? L2 NO.: 46 T A s T R A T G V F I L Q s s T N E w I S F E L R Y CDR- SEQ ID Xi? 2 X? X? X? X? X? X? X? X? X? X? L? NO.: 7 Q Q Y N N w P S - - M H N H G s L L I T Y G Y I T T P T S D Y L D C s R G S I I w V Q F I _ F F I E D. Uses of Anti-IL-18 Antibodies Given its ability to bind to human IL-18, anti-human IL-18 antibodies, or portions thereof, of the invention can be used to detect human IL-18 (e.g. , in a biological sample, such as serum or plasma), by the use of conventional immunoassay, such as enzyme linked immunosorbent assays (ELISA), a radioimmunoassay (RIA) or tissue immunohistochemistry. The invention provides a method for the detection of human IL-18 in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to the antibody. I L-1 human or unbound antibody (or antibody portion), to thereby detect human IL-18 in the biological sample. The antibody is labeled directly or indirectly with a detectable substance in order to facilitate detection of bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase or acetylcholinesterase; examples of suitable prosthetic groups include streptavidin / biotin and avidin / biotin; examples of suitable fluorescent materials include umbellifer, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinyl amine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3 H, 14 C, 35 S, 90 Y, 99 Tc, 1 l 1 ln, 25 l, 131 l, 77 Lu, 166 Ho, or 153 Sm. Alternatively to the labeling of the antibody, human I L-18 can be assayed in biological fluids by a competition immunoassay using rhlL-18 standards labeled with a detectable substance and an unlabeled anti-human IL-18 antibody. In this essay, the biological sample, the labeled rhlL-1 8 standards and the anti-human IL-18 antibody are combined and the standard amount of rhl L-1 8 labeled, bound to unlabeled antibody is determined. The amount of human I L-18 in the biological sample is inversely proportional to the amount of standard labeled rhlL-18 bound to the anti-IL-18 antibody. The antibodies and antibody portions of the invention are preferably capable of neutralizing the activity of human IL-1 8 in vitro and in vivo. Accordingly, such antibodies and antibody portions of the invention can be used to inhibit the activity of hl L-1 8, for example, in a cell culture containing hIL-18, in human subjects or in other mammalian subjects that they have I L-1 8 whereby an antibody of the invention reacts. In one embodiment, the invention provides a method for inhibiting the activity of I L-18 which comprises contacting IL-1 8 with an antibody or antibody portion of the invention in such a manner that the activity of I L-1 8 is inhibited. Preferably, IL-18 is human IL-18. For example, in a cell culture containing, or suspected of containing hIL-18, an antibody or antibody portion of the invention can be added to the culture medium to inhibit the activity of hIL-18 in the culture. In another embodiment, the invention provides a method for reducing the activity of IL-18 in a subject, advantageously from a subject suffering from a disease or disorder in which the activity of IL-18 is deleterious. The invention provides methods for reducing the activity of IL-1 in a subject suffering from such a disease or disorder, which method comprises administering to the subject an antibody or antibody portion of the invention, in such a way that the IL-18 activity in the subject. Preferably, IL-18 is human IL-18 and the subject is a human subject. Alternatively, the subject can be a mammal expressing an I L-1 to which an antibody of the invention is capable of binding. In addition, the subject can be a mammal in which hl L-18 has been introduced (for example, by administration of hl L-1 8 or by expression of a hlL-1 8 transgene). An antibody of the invention can be administered to a human subject for therapeutic purposes. In addition, an antibody of the invention can be administered to a non-human mammal expressing an IL-18 with which the antibody is capable of binding for veterinary purposes or as an animal model of human disease. With respect to the latter, such animal models may be useful for evaluating the therapeutic efficacy of the antibodies of the invention (eg, examination of doses and time courses of administration). As used herein, the term "a disorder in which the activity of I L-1 8 is harmful" is intended to include diseases and other disorders in which the presence of I L-18 in a subject suffering from the Disorder, has been shown to be or is suspected to be either responsible for the pathophysiology of the disorder or a contributing factor to a worsening of the disorder. Accordingly, a disorder in which the activity of IL-18 is harmful is a disorder in which it is expected that the reduction of the activity of I L-1 8 alleviates the symptoms and / or progress of the disorder. Such disorders may be evident, for example, by an increase in the concentration of I L-1 8 in a biological fluid of a subject suffering from the disorder (for example, an increase in the concentration of I L-1 8 in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, by the use of an anti-IL-18 antibody, as described above. Non-limiting examples of disorders that can be treated with the antibodies of the invention include those disorders discussed in the section below regarding pharmaceutical compositions of the antibodies of the invention.
D. Pharmaceutical Composition The invention also provides pharmaceutical compositions comprising an antibody, or antigen binding portion thereof, of the invention and a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition further comprises at least one additional therapeutic agent for the treatment of a disorder in which an IL-18 activity is harmful. The antibodies and antibody portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvent, dispersion media, coating, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like, which are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. The pharmaceutically acceptable carriers further comprise minor amounts of auxiliary substances such as wetting agents or emulsifiers, preservatives or regulators, which improve the shelf life or the effectiveness of the antibody or antibody portion. The antibodies and antibody portions of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration. Preferably, the antibody or antibody portions will be prepared as an injectable solution containing 0.1-250 mg / ml antibody. The injectable solution can be compounded in either a liquid or lyophilized dosage form in a flask of amber or flint, ampoule or pre-filled syringe. The regulator can be L-histidine (1-50 mM), optimally 5-10 mM, at pH 5.0 to 7.0 (optimally, pH 6.0). Other suitable regulators include, but are not limited to, sodium succinate, sodium citrate, sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, mainly 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trehalose and lactose. Volume agents can be included for a lyophilized dosage form, mainly 1-10% mannitol (optimally 2-4%). The stabilizers can be used in both liquid and lyophilized dosage forms, mainly 1-50 mM L-Methionine (optimally 5-10 mM). Other suitable bulking agents including glycerin, arginine, may be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include, but are not limited to, polysorbate 20 and BRIJ surfactants. The compositions of this invention can be found in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the proposed mode of administration and the therapeutic application. Preferred typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. The preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In a preferred embodiment, the antibody is administered by infusion or intravenous injection. In another preferred embodiment, the antibody is administered by intramuscular or subcutaneous injection. The therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome or other ordered structure, suitable for high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound (ie, antibody or antibody portion) in the required amount in a suitable solvent with one or a combination of ingredients listed above, as required, followed by filtered sterilization. In general, the dispersions are prepared by incorporating the active compound in a sterile vehicle containing a basic dispersion medium and the other ingredients required from those listed above. In the case of freeze-dried, sterile powders for the preparation of sterile injectable solutions, the preferred preparation methods are vacuum drying and spray drying which produce a powder of the active ingredient plus any additional desired ingredients from a previously filtered solution. sterile of it. The proper fluidity of a solution can be maintained, for example, by the use of a cover, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be conducted by inclusion, in the composition, of an agent delaying absorption, for example, monostearate and gelatin salts. The antibodies and antibody portions of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route / mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by those skilled in the art, the route and / or mode of administration will vary depending on the desired results. In certain embodiments, the active compound can be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are generally patented or known to those skilled in the art. See, for example, Delivery Systems for Prolonged and Controlled Release Drug, J. R. Robinson, ed. , Marcel Dekker, Inc., New Cork, 1978. In certain embodiments, an antibody or antibody portion of the invention can be administered orally, for example, with an inert diluent or an edible assimilable carrier. The compound (and other ingredients, if desired) can also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or directly incorporated into the subject's diet. For oral therapeutic administration, the compounds can be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, dragees, capsules, elixirs, suspensions, syrups, wafers and the like. To administer a compound of the invention by administration other than parenteral, it may be necessary to coat the compound with, or co-administer the compound with, a material in order to prevent its inactivation. The complementary active compounds can also be incorporated into the compositions. In certain embodiments, an antibody or antibody portion of the invention is co-formulated with and / or co-administered with one or more additional therapeutic agents that are useful for the treatment of disorders in which the activity of I L is harmful. -18. For example, an antibody or anti-hIL-8 antibody portion of the invention can be co-formulated and / or co-administered with one or more additional antibodies that bind to other targets (eg, antibodies that bind to other cytokines or that bind to cell surface molecules). In addition, one or more antibodies of the invention can be used in combination with two or more of the above therapeutic agents. Such combination therapies can advantageously use lower doses of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. In certain embodiments, an antibody to IL-1 or fragment thereof is linked to a medium-lived expander vehicle known in the art. Such carriers include, but are not limited to, the Fe domain, polyethylene glycol, and dextran. Such vehicles are described, for example, in the Application of E. U. Series No. 09/428, 082 and PCT Application published No. WO 99/25044, which is incorporated herein by reference for any purpose. Interleukin 1 8 plays a critical role in the pathology associated with a variety of diseases involving immune and inflammatory elements. These diseases include, but are not limited to, rheumatoid arthritis, osteoarthritids, juvenile chronic arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory liver disease, insulin-dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, scleroderma dermatitis, graft-versus-host disease, rejection of organ transplantation, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener granulomatosis, Henoch-Schoenlein purpura, microscopic vasculitis of the kidneys, chronic active hepatitis, uvitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, Infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, transverse myelitis to acute, Huntington's disease, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, type I polyglandular deficiency, and type II polyglandular deficiency , Schmidt syndrome, respiratory dysfunction syndrome in adults (acute), alopecia, alopecia areata, seronegative artopathy, arthropathy, Reiter's disease, psoriatic artopathy, ulcerative eolithic artopathy, enteropathic synovitis, chlamydia, arthropathy associated with yersinia and salmonella, spondyloartopathy, atheromatous disease / arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, penphigus foliaceus, pemphigoid, linear IgA disease, autoimmune hemolytic anemia, positive hemolytic anemia of Coombs, acquired pernicious anemia, juvenile pernicious anemia, myalgic encephalitis / Royal Free disease , mucocutaneous candidiasis chronic, giant cell arteritis, primary sclerosing hepatitis, autoimmune hepatitis Diseases Related to Acquired Immunodeficiency, Hepatitis B, Hepatitis C, common variable immunodeficiency (common variable hypogammaglobulinemia) ', dilated cardiomyopathy, female infertility, ovarian failure, premature failure of ovaries, disease fibrotic puionar, critypelenic fiber alveoliths, post-inflammatory interstitial lung disease, interstitial pneumonitis, interstitial lung disease associated with connective tissue disease, lung disease associated with mixed connective tissue disease, instersticial lung disease associated with systemic sclerosis, associated interstitial lung disease with rheumatoid arthritis, pulmonary disease associated with systemic lupus erythematosus, pulmonary disease associated with dermatomyositis / polymyositis, lung disease associated with Sjögren's disease, lung disease associated with ankylosing spondylitis, diffuse vasculitic pulmonary disease, lung disease associated with hemosiderosis, drug-induced instersticial lung disease, fibrosis, radiation fibrosis, obliterans bronchiolitis, chronic eosinophilic pneumonia, lymphocytic infiltrative pulmonary disease, post-infectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, autoimmune hepatitis type-1 (classic or lupoid autoimmune hepatitis), autoimmune hepatitis type 2 (hepatitis of anti-LKM antibodies), autoimmune mediated hypoglycemia, insulin resistance type B with nygranin acanthosis, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, type 2 psoriasis, idiomatic leukopenia, autoimmune neutropenia, NOS of kidney disease, glomerulonephritis, microscopic vasculitis of the kidneys, slime disease, discoid lupus erythematosus, male language infertility or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), ophthalmia sympathetic, pulmonary hypertension secondary to connective tissue disease, Goodpasture syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjórgren's syndrome, Takayasu's disease / arteritis, autoimmune thrombocytopenia, idiomatic thrombocytopenia , autoimmune thyroid disease, hyperthyroidism, autoimmune hypothyroidism with goiter (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uvitis, primary vasculitis, acute liver disease vitiligo, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver damage, coleosatatis, idiosyncratic liver disease, drug-induced hepatitis, non-alcoholic steatohepatitis, allergy and asthma, group B streptococcal (GBS) infection, mental disorders (eg, depression and schizophrenia), Type Th2 and Type Th1, acute and chronic pain (different forms of pain), and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovaries, prostate, and rectal cancer and hematopoeic malignancies (leukemia and lymphoma) . The human antibodies and the antibody portions of the invention can be used to treat humans suffering from autoimmune diseases, in particular those associated with inflammation, including rheumatoid spondylitis, allergy, autoimmune diabetes, autoimmune uvitis. Preferably, the antibodies of the invention or antigen binding portions thereof, are used to treat rheumatoid arthritis, Crohn's disease, multiple sclerosis, insulin-dependent diabetes, melitus and psoriasis. An antibody or antibody portion of the invention can also be administered with one or more additional therapeutic agents, useful in the treatment of autoimmune and inflammatory diseases. The antibodies of the invention or antigen binding portions thereof can be used alone or in combination to treat such diseases. It is to be understood that the antibodies of the invention, or antigen binding portion thereof, can be used alone or in combination with an additional agent, for example, a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent may be a therapeutic agent recognized in the art to be useful in treating the disease or condition to be treated by the antibody of the present invention. The additional agent can also be an agent imparting a beneficial attribute to the therapeutic composition, for example, an agent that affects the viscosity of the composition.
It should further be understood that the combinations to be included within this invention are those combinations useful for their intended purpose. The agents stated below are for illustrative purposes and do not intend to limit themselves. The combinations, which are part of the invention, may be the antibodies of the present invention and at least one additional agent selected from the lists below. The combination may also include more than one additional agent, for example, two or three additional agents if the combination is such that the formed composition can carry out its intended function. Preferred combinations with non-spheroidal anti-inflammatory drug (s) also referred to as NSAIDS include drugs such as ibuprofen. Other preferred combinations with corticosteroids including prednisolone; the well-known side effects of the use of steroids can be reduced or even eliminated by tapering the required steroid dose when treating patients in combination with the anti-IL-18 antibodies of this invention. Non-limiting examples of therapeutics for rheumatoid arthritis with which an antibody or antibody portion of the invention can be combined include the following: cytosine suppressor (s) anti-inflammatory drug (s) (CSAIDs); antibodies or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 , IL-12, IL-15, IL-16, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD28, CD28, CD30, CD40, CD45, CD69, CD80 (B7) .1), CD86 (B7.2), CD90, CTLA or its ligands including CD154 (gp39 or CD40L). Preferred combinations of therapeutic agents may interfere at different points in the subsequent autoimmune and inflammatory cascade; preferred examples include TNF antagonists such as chimeric, humanized or human TNF antibodies, D2E7, (PCT Application No. WO 97/29131), CA2 (Remicade ™), CDP571, and soluble p55 or p75 TNF receptors, derivatives thereof, (p75TNFR1 gG (Enbrel ™) or p55TNFR1 gG (Lenercept) and also inhibitors of the TNFa conversion enzyme (TACE); inhibitors I L-1 similarly (inhibitors of enzyme conversion to lterleukin-1, 1L-1 RA, etc.) may be effective for the same reason Other preferred combinations include Iterleukin 11 1. Still another preferred combination are other key players of the autoimmune response that can act parallel to, dependent on or in concert with IL-1 function. Especially preferred are IL-12 antagonists which include I L-12 antibodies or soluble I L-12 receptors, or IL-12 binding proteins.It has been shown that IL-12 and IL-18 have different functions but that they overlap and a combination of antagonists for both you can be the most effective. Still another preferred combination are the non-depleting anti-CD4 inhibitors. Still other preferred combinations include antagonists of the costimulatory pathway CD80 (B7.1) or CD86 (B7.2) which includes antibodies, soluble receptors or antagonistic ligands. Antibodies of the invention, or antigen binding portions thereof, may also be combined with agents such as methotrexate, 6-MP, azathioprine sulfasalazine, mesalazine, olsalazine chloroquine / hydroxychloroquine, pencitamine, aurothiomalate (intramuscular and oral), azathioprine, coquicina, corticosteroids (oral, inhaled and local injection), beta-2-adrenoreceptor agonists (salbutamol, ttaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, quetotifen, ipratropium and oxitropium, ciclosporin, FK506, rapamycin , mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents that interfere with signaling by pro-inflammatory cytokines such as TNFa or I L-1 (e.g., I RAK, NIK, I KK, p38 or MAP kinase inhibitors), I L-1β conversion enzyme inhibitors, TNFα conversion enzyme inhibitors (TACE), T cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6- mercaptopurins, angiotensin-converting enzyme inhibitors, soluble cytosine receptors and derivatives thereof (for example, soluble p55 or p75 TNF receptors and the p75TNFR1 gG derivatives (Enbrel ™ and p55TNFRIgG (Lenercept), slL-1 Rl, slL -1 Rll, slL-6R), anti-inflammatory cytokines (for example, I L-4, IL-10, IL-1 1, IL-1 3 and TGFβ) celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab , napro xeno, valdecoxib, sulfasalizine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium thiomálate, aspirin, triamcinolone acetonide, propoxyphene napsilate / apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone hcl , hydrocodone bitartrate / apap, sodium diclofenac / misoprostol, fentanyl, anakinra, human recombinant, hcl tramadol, 'salsalate, sulindac, cyanocobalamin / fa / pyridoxine, acetaminophen, alendronate sodium, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, sulfosaline / chondroitin, hcl amitriptyline, sulfadiazine, hcl oxycodone / acetaminophen, hcl olopatadine, misoprostol, naproxen sodium, omeprazole, cyclophosphamide, rituximab, I L-1 TRAP, MRA, CTLA4-IG, IL-1 8 BP, anti -I L-12, anti-I L1 5, BIRB-796, SCIO-469, SVX-702, AMG-548, VX-740, Roflumilast, IC-485, CDC-801, and Mesopram. Preferred combinations include methotrexate or leflunomide and in cases of moderate or severe rheumatoid arthritis, cyclosporin. Non-limiting examples of therapeutic agents for inflammatory liver disease with which an antibody, or antibody portion of the invention can be combined, include the following: budenoside, epithelial growth factor; corticosteroids; cyclosporin, sulfasalazine; amiosalicylates; 6- mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balzalazide; antioxidants; thromboxane inhibitors; receptor antagonists of 1 L-1; anti-IL-1β monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl imidazole compounds; antibodies or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, I L-7, IL-8, I L-12, I L-15, I L-16, EMAP-I I, GM-CSF, FGF and PDGF. The antibodies of the invention or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD28, CD28, CD40, CD45, CD69, CD69, or their ligands. . Antibodies of the invention or antigen binding portions thereof, can also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, eg, ibuprofen, corticosteroids such as prednisolone, inhibitors. of phosphodiesterase, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents that interfere with the signaling of proinflammatory cytokines such as TNFa or I L-1 (for example, I RAK, NI K. I KK, p38 or inhibitors of MAP kinase), IL-1β conversion enzyme inhibitors, TNFα conversion enzyme inhibitors, T cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, enzyme inhibitors of Angiotensin conversion, soluble cytosine receptors and derivatives thereof (e.g., TNF receptors of p55 or p75 so lubles, slL-1 Rl, if L-1 Rl I, sl L-6R) and anti-inflammatory cytokines (for example, IL-4, IL-10, I L-1 1, IL-13 and TGFβ). Preferred examples of therapeutic agents for Crohn's disease in which an antibody or an antigen binding portion can be combined include the following: TNF antagonists, eg, anti-TNF antibodies, D2E7 (PCT Application No. WO 97) / 29131; HUMRA), CA2 (REMICADE), CDP 571, TNFR-lg constructs, inhibitors (p75TNFRIgG (ENBREL) and p55TNFRIgG '(LERNERCEPT) and PDE4 inhibitors.The antibodies of the invention, or antigen binding portions of they can be combined with corticosteroids, for example, budenoside and dexamethasone Antibodies of the invention or antigen binding portions thereof, can also be combined with agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine, and interfering agents. with the synthesis or action of pro-inflammatory cytokines such as IL-1, for example, inhibitors of conversion enzyme l L-1β and IL-1 ra The antibodies of the invention or binding moiety antigen thereof can also be used with T cell signaling inhibitors, for example, 6-mercaptopurines of tyrosine kinase inhibitors. The antibodies of the invention or antigen binding portion thereof can be combined with IL-1 1. Antibodies of the invention or antigen binding portion thereof can be combined with mesalamine, prednisone, azathioprine, mercaptopurine, infliximab, methylprednisolone sodium succinate, diphenoxylate / atrop sulfate, loperamide hydrochloride, methotrexate, omeprazole, folate, ciprofloxacin / dextrose-water, hydrocodone bitartrate / apap, tetracycline hydrochloride, fluocinonide, metronidazole, thimerosal / boric acid, cholestyramine / sucrose, ciprofloxacin hydrochloride, hyoscyamine sulfate, meperidine hydrochloride, midazolam hydrochloride, oxycodone / acetaminophen hcl, hydrochloride promethazine, sodium phosphate, sulfamethoxazole / trimethoprim, celecoxib, polycarbophil, propoxyphene napsylate, hydrocortisone, multivitamins, balsalazide disodium, codeine / apa phosphate, hcl colesevelam, cyanocobalamin, folic acid, levofloxacin, methylprednisolone, natalizumab and interferon-gamma. Non-limiting examples of therapeutic agents for multiple sclerosis with which an antibody, or antibody portion of the invention can be combined, include the following: corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporin; methotrexate; 4-aminopyridine; tizanidine; interferon-ß1 a (AVONEX; Biogen); interferon-β1 b (BETASERON; Chiron / Berlex); interferon a-n3) (Interferon Sciences / Fujimoto), interferon-a (Alpha Wassermann / J &J), interferon ß1 AI F (Serono / Inhale Therapeutics), Peginterferon a 2b (Enzon / Schering-Plow), Copolymer 1 ( Cop-1; COPAXONE; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; Clabribine; antibodies or antagonists of other human cytokines or growth factors and their receptors, for example, TNF, LT, I L-1, I L-2, IL-6, I L-7, I L-8, I L-12 , IL-23, I L-15, I L-16, EMAP-I I, GM-CSF, FGF and PDGF. Antibodies of the invention or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD20, CD20, CD28, CD28, CD40, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands. Antibodies of the invention or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomine, NSAIDs, eg, ibuprofen, corticosteroids such as prednisolone, inhibitors. of phosphodiesterase, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents that interfere with signaling by pro-inflammatory cytokines such as TNFa or I L-1 (e.g., IRAK, NIK, I KK, kinase inhibitors p38 or MAP), I L-1β conversion enzyme inhibitors, TACE inhibitors, T cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin-converting enzyme inhibitors , soluble cytosine receptors and derivatives thereof (e.g., soluble p55 or p75 TNF receptors, sl L-1 R l, if L-1 Rl l, sl L-6R) and anti-inflammatory cytokines (e.g., IL-4, IL-10, IL-13 and TGFβ). Preferred examples of therapeutic agents for multiple sclerosis in which the antibody or antigen binding portion thereof can be combined to include interferon-β, for example, IFNßl a and IFNßl b; copaxone; corticosteroids, caspase inhibitors, e.g., caspase-1 inhibitors, IL-1 inhibitors, TNF inhibitors, and antibodies to CD40 and CD80 ligands. Antibodies of the invention or antigen binding portions thereof can also be combined with agents such as alemtuzumab, dronabinol, Unimed, daclizumab, mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunoquine, NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2278, calagualin, CPI-1 189, LEM (mitoxantrone encapsulated by liposome), THC.CBD (cannabinoid agonist) MBP-8298, mesopram (PDE4 inhibitor), MNA-715, anti-I receptor antibody L-6, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-RI, talampanel, teriflunomide, TGF-beta2, tiplimotide, VLA-4 antagonists (by example, TR-14035, VLA4 Ultrahaler, Integran-ELAN / Biogen), interferon gamma antagonists, IL-4 agonists. Non-limiting examples of therapeutics for Angina with which an antibody, or antibody portion, of the invention can be combined, include the following: aspirin, nitroglycerin, isosorbide mononitrate, metoprolol succinate, atenolol, metoprolol tartrate, besylate amlidipine, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, hcl verapamil, digoxin, propranolol hydrochloride, carvedilol, lisinopril, spironolactone, hydrochlorothiazide, enalapril maleate, nadolol , ramipril, enoxaparin sodium, heparin sodium, valsartan, sotalol hydrochloride, fenofibrate, ezetimibe, bumetanide, losartan potassium, lisinopril / hydrochlorothiazide, felodipine, captopril, bisoprolol fumarao. Non-limiting examples of therapeutic agents for Ankylosing spondylitis with which an antibody or antibody portion of the invention may be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, sulfasalazine, methotrexate, azathioprine, minocycline, prednisone, etanercept, infliximab Non-limiting examples of therapeutics for Asthma with which an antibody or antibody portion of the invention can be combined include the following: albuterol, salmeterol / fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, hcl levalbuterol, albuterol / ipratropium sulphate, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pyrbuterol acetate, prednisolone, anhydrous theophylline, methylprednisolone sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin thihydrate, flunisolide, allergy shot, cromolyn sodium, fexofenadine hydrochloride, flunisolide / menthol, amoxicillin / clavulanate, levofloxacin, ancillary device inhaler, guaifenesin, sodium dexamethasone phosphate, hcl moxifloxacin, doxycycline hyclate, guaifenesin / d-metorf an, p-ephedrine / cod / chlorfenir, gatifloxacin, cetirizine hydrochloride, mometasone furoate, salmeterol xinafoate, benzonatate, cephalexin, pe / hydrocodone / chlorphenir, hcl cetirizine / pseudoefed, phenylephrine / cod / promethazine, codeine / promethazine, cefprozil , dexamethasone, guaifenesin / pseudoephedrine, chlorpheniramine / hydrocodone, nedocromil sodium, terbutaline sulfate, epinephrine, methylprednisolone, metaproterenol sulfate. Non-limiting examples of therapeutic agents for COPD with which an antibody or antibody portion of the invention can be combined include the following: albuterol sulfate / ipratropium, ipratropium bromide, salmeterol / fluticasone, albuterol, salmeterol xinafoate, fluticasone propionate, prednisone, anhydrous theophylline, sodium succinate of methylprednisolone, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, hcl levalbuterol, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin / clavulanate, flunisolide menthol, chlorpheniramine / hydrocodone, metaproterenol sulfate, methylprednisolone, mometasone furoate, p-ephedrine / cod / chlorfenir, pyrbuterol acetate, p-ephedrine / loratadine, terbutaline sulfate, tiotropium bromide, (R, R) -formoterol, TgAAT, Cilomilast, Roflumilast. Non-limiting examples of therapeutic agents for HCV with which an antibody or antibody portion of the invention can be combined include the following: Interferon-alpha-2a, Interferon-alpha-2b, Interferon-alpha with l, lterferon-alpha-n 1, Pegylated interferon-alpha-2a, Interferon-alpha-2b Pegylated, ribavirin, Peginterferon alfa-2b + ribavirin, Ursodeoxycholic acid, Glycyrrhizic acid , Timalfasin, Maxamine, VX-497 and any compound that is used to treat HCV through intervention with the following objectives: HCV polymerase, HCV protease, HCV helicase, HCV I RES (internal ribosome entry site) . Non-limiting examples of therapeutic agents for Idiomatic Pulmonary Fibrosis with which an antibody or antibody portion of the invention can be combined include the following: prednisone, azathioprine, albuterol, colchicine, albuterol sulfate, digoxin, gamma interferon, methylprednisolone succ sod, lorazepam, furosemide, lisinopril, nitroglycerin , spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, hcl oxycodone, potassium chloride, triamcinolone acetonide, anhydrous tacrolimus, calcium, interferon-alpha, methotrexate, Mycophenolate mofetil, Interferon-gamma-1 ß. Non-limiting examples of therapeutic agents for Myocardial Infarction with which an antibody or antibody portion of the invention can be combined include the following: aspirin, nitroglycerin, metoprolol tartrate, enoxaparin sodium, heparin sodium, clopidogrel bisulfate, carvedilol , atenolol, morphine sulfate, metoprolol succinate, sodium warfarin, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril hcl / carb mag, bumetanide , alteplase, enalaprilat, amiodarone hydrochloride, hcl tirofiban m-hydrate, diltiazem hydrochloride, captopril, irbesartan, valsaran, propranolol hydrochloride, fosinopril sodium, lidocaine hydrochloride, eptifibatide, cefazolin sodium, atropine sulfate, aminocaproic acid, spironolactone, interferon, sotalol hydrochloride, potassium chloride, docusate sodium, dobutamine hcl na, alprazolam, pravastain sodium, aiovasaline calcium, midazolam hydrochloride, meperidine hydrochloride, isosorbide dinitrate epinephrine, dopamine hydrochloride, bivalirudin, rosuvastatin, ezetimibe / simvastatin, avasimibe, cariporide. The non-limiting examples of self-regulatory agencies for Psoriasis with which an antibody or antibody portion of the invention can be combined include the following: calcipotriene, clobetasol propionate, triamcinolone acetonide, halobetasol propionate, iazaroin, meiotrexate, fluocinonide, increased betamethasone diprop, fluocinolone acetonide, acitretin, tar shampoo, betameiasone valerate, momefasone furoa, quenoconazole, pramoxin / fluocinolone, hydrocorisone valerate, flurandrenolide, urea, beiameiason, clobeiasol propionate / emol, fluioicasone propionate, azithromycin, hydrocorisone, humeciole formula, folic acid, desonide, pimecrolimus, vegetable charcoal alkylamine, diflorasone diaceia, eianercepie folate, lactic acid, meioxsalen, bismuth / znox subalgal / resor, meilyprednisolone acetyl, prednisone, solar procerr, halcinonide, salicylic acid, aniraline, clovinylolone pivalate, Extraction of charcoal, tar charcoal / salicylic acid, vegetable charcoal / salicylic acid / sulfur, deoxymethiasone, diazepam, emollient, fluocinonide / emollient, mineral oil / castor oil / nacium, mineral oil / peanut oil, peiroleum / isopropyl myrosium , psoralen, salicylic acid, soap / fribromsalan, thimerosal / boric acid, celecoxib, infliximab, cyclosporine, alefacept, efalizumab, iaccrolimus, pimecrolimus, PUVA, UVB, sulfasalazine. Non-limiting examples of peptide agents for psoriatic arthritis with which an antibody or antibody portion of the invention can be combined include the following: methotrexate, etanercept, rofecoxib, celecoxib, folic acid, sulfasalazine, naproxen, leflunomide, methylprednisolone acrylate, indomethacin , hydroxychloroquine sulfate, prednisone, sulindac, increased betameiasone dipropyl, infliximab, meioirexine, folate, iodiamcinolone acetylide, diclofenac, dimefilsulfoxide, piroxicam, diclofenac sodium, aceoprophen, meloxicam, methylprednisolone, nabumetone, tolmein sodium, calcipotriene, cyclosporine, diclofenac sodium / misoprostol, fluocinonide, glucosaline sulfate, gold sodium iodide, hydrocodone / apap biomarine, ibuprofen, risedronate sodium, sulfadiazine, ioguanine, valdecoxib, alefacepí, efalizumab. Non-limiting examples of therapeutic agents for restenosis with which an antibody or antibody portion of the invention can be combined include the following: sirolimus, paclitaxel, everolimus, tacrolide us, ABT-578, aceiaminophen. Non-limiting examples of psychoactive agents for Cassia with which an antibody or antibody portion of the invention can be combined include the following: hydrocodone / apap biomarine, rofecoxib, cyclobenzaprine hcl, meilprednisolone, naproxen, ibuprofen, oxycodone / acetylaminophen, celecoxib, valdecoxib, meilyprednisolone acetylation, prednisone, codeine / apap phosphation, hramaladol / acetaminophen, etaxalone, meloxicam, medeocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapenin, dexamethasone, carisoprodol, queyorolac meramylamine, indomeiacin, aceiaminophen, diazepam , nabumetone, oxycodone, hcl tizanidine, diclofenac sodium / misoprostol, propoxyphene napsilay / apa, asa / oxycodone / oxycodone, ibuprofen / hydrocodone bi, hcldramadol, edolac, hcl propoxyphene, hcl amltriptiline, carisoprodol / codeine fos / asa , morphine sulfate, multivitamins, sodium of naproxen, citraio of orphenadrine, íemazepam . Preferred examples of idiopathic agents for SLE (Lupus) in which an antibody or antibody portion of the invention can be combined include the following: NSAIDs, eg, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example Celecoxib, rofecoxib, valdecoxib; ani-malaria, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexameriasone; Ciioioxics, for example, azaíioprina, cyclophosphamide, mycophenolate myophenolate, methotrexium; PDE4 inhibitors or purine synisis inhibitor, e.g., Cellcept. Antibodies of the invention or antigen binding portions thereof can also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents that interfere with the synthesis, production or action of pro-inflammatory cytokines such as I L-1, for example, caspase inhibitors as inhibitors of IL-1β and I L-1 ra conversion enzyme. The antibodies of the invention or antigen binding portions thereof can also be used with inhibitors of T cell signaling, eg, tyrosine kinase inhibitors; or molecules that direct the T cell activation molecules, for example, CTLA-4-IgG or antibodies of the anti-B7 family, antibodies of the anti-PD-1 family. The antibodies of the invention or antigen binding portions thereof can be combined with I L-1 1 or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg antibody), or host receptor-receptor antibodies, for example. , receptor antibody of anti-IL-6 and antibodies to B-cell surface molecules. Antibodies of the invention or antigen-binding portions thereof can also be used with LJP 394 (abetimus), agents that deplete or inactivate B cells. , for example, Rimoximab (ani-antibody ani-CD20), lymphosia-B (aniibody anfi-BlyS), aniagonisías TNF, for example, aniibodies aníi-TNF, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 ( REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL) and p55TNFRIgG (LENERCEPT)). The pharmaceutical compositions of the invention may include a "therapeutically effective amount" or a "prophylactically effective amount" of an antibody or antibody portion of the invention. An "ineffective effective amount" refers to an effective amount, at doses and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may be determined by a person experienced in malaria and may vary according to natural factors such as disease status, age, sex and weight of the individual and the ability of the antibody or antibody portion to produce a desired response in the individual. A highly effective epistemic mass is also one in which any toxic or noxious effect of the antibody or antibody portion would be desired through the beneficial effects of the disease. A "most effective prophylactic principle" refers to an effective frequency, at doses and for necessary periods of time, to achieve the desired prophylactic outcome. Typically, since a prophylactic dose is used in fixations from or in an early stage of the disease, the most effective prophylactic frequency will be less than the most effective feperalicy canine. Dosage regimens can be adjusted to provide the desired optimal response (for example, an idiopathic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered at the same time, or the dose may be proportionally reduced or increased as indicated by the requirements of the idiographic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form to facilitate administration and uniformity of dosage. The dosage unit form, as used herein, refers to physically discrete units, suitable as unit doses for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound, calculated in order to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification of dosage unit forms of the invention is dictated by and directly dependent on (a) the unique characteristics of the active compound and the eedemic effect or prophylactic effect to be achieved., and (b) the inherent limitations in the composition composition of the active compound for the sensitivity trait in individuals. An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody or portion of the antibody of the invention is 0.1-20 mg / kg, more preferably 1 -1.0 mg / kg. It should also be understood that for any specific device, the specific dose regimens should be adjusted with time according to the individual need and professional judgment of the person administering or supervising the administration of the compositions and dose ranges established in the present are only exemplary and are not intended to limit the scope or practice of the claimed composition.
II. Responsive genes of IL-18 I L-18 is expressed in macrophages, dendritic cells, Kupffer cells, microglia, epiíeliales cells, keraíinociíos, inifinal epifelial cells, chondrocytes, synovial fibroblasts and osteoblasts, as well as within the adrenal cortex and pituitary gland . In some cells, such as human monocytes and dendritic cells, the expression is consistent, while in other cells it must be induced again. Apare of the gamma expression of inerferona, little is known about other genes induced by IL-18 alone or in relation to other cytosomes. One embodiment of the invention provides a method for regulating the gene expression of an immune gene comprising the steps of providing L-1 8 or a modulator I L-18; and containg I-L-18 or the modulator to a cell wherein the gene of genesis is selected from the group consisting of the genes shown in the following table.
Table 3 Receptive genes IL-18 Inhibitory currents of I L-1 8 are described, for example, in the USA Patent. No. 5,912, 324, October 14, 1994; EP 0 962 531, published on December 8, 1999; EP 712 931, published November 15, 1994; Pai. from USA UU No. 5,914,253, issued on July 4, 1994; WO 97/24441, published July 10, 1997; Pai. from the USA No.6, 060, 283, issued on May 9, 2000; EP 850 952, published December 26, 1996; EP 864 585 published on September 16, 1998; WO 98/41232, published on Sep. 24, 1998; Pat. from USA UU No.6, 054,487, granted on April 25, 2000; WO 99/09063, published on August 14, 1997; WO 99/22760, published November 3, 1997; WO 99/37772, published January 23, 1998; WO 99/37773, published March 20, 1998; EP 0 974 600, published on January 26, 2000; WO 001 12555, published March 9, 2000; Japanese Patent Application JP 1 1 1, 399, 194, published on 31 October 1997; request of Israel pa.l. IL 121 554 A0, published on February 8, 1998; which are incorporated in the present for reference for any purpose. It will be readily apparent to those skilled in the art that other suitable modifications and adaptations of the methods of the invention described herein are obvious and may be elaborated using suitable equivalents without departing from the scope of the invention or the embodiments described herein. Having now described the present invention in detail, it will be more readily understood for reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting of the invention.
EXAMPLES Example 1: Production and Characterization of recombinant IL-18 Example 1.1: Assays for determining biological activity of 1L-18 Throughout Example 1 and Example 2 the following assays were used to determine biological activity of I L-18 unless stated otherwise.
Example 1.1. A: Bioensavo KG-1 KG-1 (ATCC # CCL-246) is a human myelomonocytic cell strain expressing low levels of functional IL-18 receptor. Treatment with TNF regulates both the IL-18Ra and ß subunits of the functional L-18 receptor in these cells. The KG-1 bioassay was performed by incubating KG-1 cells treated by TFN with recombinant human L-18 (rhu-IL-18) and determining the level of IFNα production. induced by 1L-1 8 by an ELISA (Konishi, K., ef al. (1997) J. Immunol. Methods 209: 187-1 91). The KG-1 bioassay was used to determine the potency of oligonazation 1-18. For example, the L-18 aniibodies were incubated with different concentrations of rhu-IL-18 and incubated in this way with KG-1 cells treated with TNF in a 96-well plate for 16-18 hours at 37 ° C. The supernatants were tested and collected for IFNα levels. human by an ELISA. This assay can measure the IC50 values down to 4x1 0"1 1 6 x10" 1 1M of an anonymysia I L-1 8.
Example 1.1. B: Human Complete Blood Test In summary, the Human Complement Blood Test (WBA) demystifies the neu- ralization of IL-18 antipharyngeal coníra with a naïve IL-18 from a physiological coníexy. In this trial, the lecture was the inhibition of IFNα production. Human endogenous 1-1 8 dependent. Whole blood was stimulated with LPS (1 μg / ml) plus IL-12 (50 pg / ml) in the presence or absence of antagonisms I L-18 at 37 ° C. Concentrations of IFN? were determined by ELISA 18-24 hrs posi-LPS plus IL-12 eslimulation.
Example 1.1. C: Receptor Binding Assay In summary, the Receptor Binding Assay (RBA), 125 l labeled rhu-IL-1 8 was used to determine the binding of I L-8 to the receptacle I L-1 8. 125l-rhu -IL-18 specifically binds to IL-1 8Raβ in KG-1 cells irradiated by TNF (-7,000 cells / cells). 125l-rhu-IL-18 has the same specific activity as unlabeled IL-18 and can be competed for unlabeled I L-1 8. Two modes of inhibition, A and B, were defined. In Mode A neutralization, the binding of IL-18 to the high affinity IL-18 receptor (IL-18Raβ) was not effected, but the signal transduction mediated by I L-18 (ie, IFNα production) it's blocked. In Neutralization Mode B, the binding of I L-18 to I L-1 8Raß was blocked and thus receptor-mediated signaling did not occur.
Example 1.2: Production of recombinant IL-18 Example 1.2. A: Plasmid construction. Expression and purification of human prolL-18 I recombinant human L-18 was generated by expressing the precursor form of L-1 8 in SF-9 insect cells. Using the standard molecular biological methods well known in matheria, full-length human cDNA pro-lL-1 was generated using PCR primers based on the published sequence (Ushio S. et al. (1 996) J. Immunol. : 4274-4279) and was subsequently cloned into the baculovirus (BV) transfer vector pVI1 393. (BD Biosciences, San Jose, CA; Cat # 51 -21201 P). The 5'PCR primer used to generate the human long-term pro-IL-1 8 cDNA complemented with sequences coding for the 6-Hisiinde region in such a way that the N-terminal of the pro-I L-1 8 produced a 6-HIS effect. . The SF9 insemination cells were infected with the neighbor pVL1393 which hosts baculovirus conferring IL-18 cDNA. The SF9 cells harvested were lysed by lysines and the lyses run on a nickel column to purify pro-IL-18 labeled HIS recombinant (rhu pro-I L-1 8). (BD Biosciences, San Jose, CA; Cat. # 554802). Pro-IL-18 labeled recombinant HIS was further processed by being digested with human caspase-1 to generate biologically active 1L-18 (mature I-L-18). (Ghayur T., (1997) Nature 386: 619-623).
Example 1.2.B: NEM treatment of recombinant human IL-18 IL-18 obtained from Hayashibara Biochemical Laboratories, Japan, showed the variation of group in specific activity and binding affinity I L-1 8. IL-18 contained disulfide bonds between several pairs of the four cisterns in mature IL-18. These originated functional and structural heterogeneity, and variations between groups. The homologous molding of human L-1 8 using the coordinates IL-1 b showed that the cysteine residues at positions 38 and 68 of mature human L-18 are exposed and therefore are reactive. I Recombinant human L-18 of Example 1 .2. A was added with N-methyl maleiamide (NEM) to provide the oxidation cistrols. NEM-I L-1 8 was monomeric, did not form aggregates, was stable, and retained high specific activity over time. NEM-IL-18 retained the neutralizing epitopes because the neutralizing anti-hul L-18 antibodies bound and neutralized NEM-IL-1 8. Instead of the NEM tramitame of I L-18, the neutralizing epííopos in NEM-I L-18 were conserved as determined by the ability of anti-IL-18 antibodies to neutralize the biological activity of NEM-IL-18 as human naive IL-18 in human WBA. NEM-IL-18 was used for the optimization of assay and selection and initial characterization of anti-human or completely human IL-18 mAbs.
Example 1.2.C: Generation and characterization of IL-18 mutant 4C / A The mutant 4C / A of IL-18 was generated by mutating the four Cysteine residues in mature IL-18 for Alanine ("4C / A-huL- 18"). Comparison of mutant 4C / A of I L-18 with NEM-hulL-18, as summarized in Table 4 below, showed that the two proieins were indistinguishable in biochemical and biological properties. Both 4C / A mutant of IL-18 and NEM-hul L-1 8 were monomeric by dynamic light scattering (DLS) and size exclusion chromatography (SEC) analysis, and similar in conformation and physical stability by dichroism analysis circular. The biological activity of 4C / A mutant and NEM-hulL-1 8 were the same in the KG-1 assay, and both forms of I L-18 bound IL-18BP and ani-I antibodies L-18 with similar affinity. 4C / A-huL-18 was not subject to oxidative instability and was easily expressed at alios levels in E. coli.
Table 4 The comparison between NEM-IL-18 and 4C / A-hulL-18 shows that they are equivalent in measurements of conformational and oligomeric purity, physical stability, binding to antibodies or cell-bound receptors.
Oligomeric State CD (CD length minimum to 210 CD minimum at 210 Confirmation wave scanned) nm nm CD (Stable temperature above Stable above Scanned stability) 40 ° C 40 ° C I FNy production Bioactivity of 2 ng / mL IL-1 8 8 ng / mL IFNY 8 ng / mL IFNY Neutralization of Neutralized by Neutralized by IFNy production IL-1 8BP-Fe, 125-IL-18BP-Fe, Epitopes by binding 2H and IL-l8Ra 1252H and IL-18Ra reference 1L-1 8BP-Fc: IL-18BP-Fc: 0.098 0. 135 nM 125-2H: 0.2 Biacore (KD) 125-2H: 0.2 nM nM 125-2H: 0.2 2.5 (E) mg 1: 0.3 nM nM Example 1.2.D: Generation and characterization of biotinylated rhu-IL-18 (biot-IL-18, Bioinj NEM-I L-18 of Example 1 .2.B was performed on lysine residues using standard techniques in the art, (Sulfo-NHS-LC-Bioin, Pierce, Rockford, IL; #Cat 21335), and rhu-IL -18 biotinylated (biot-IL-1 8) was a heterogeneous mixture that contains a species with 1, 2, 3, or 4 bioins per hulL-1 8. In addition, the species with 2 or 3bioinines per rhulL-18 were major species in biot-I L-1 8. BioI-IL-1 8 was biologically active, anti-IL-18 antibodies bound as determined by ELISA, and neutralized by all tested anti-hulL-18 neutralizing antibodies. -1 bound by BioI-IL-18 expressing IL-18Raβ on its surface with alia affinity, and bio-IL-18 on the surface of KG-1 cells was determined by FAC analysis using anti-biotin antibodies (Sigma-Aldri ch, Yes. Louís, MO; # fell, B 3640). In this manner, bioininylation does not arise with receptor binding and does not mask the neutralizing epitopes of rhulL-18.
Example 1.2.E: Generation and characterization of rhulL-18 labeled 125l The lysine residues in NEM-IL-18 of Example 1 .2. B were labeled with 125 I using conditions specified by Amersham (Piscataway, NJ; # Cat. IM5861). I L-18 labeled 125I retained its specific activity, was com- bined by unmodified IL-18, and specifically bound to I L-18R in KG-1 cells. The binding of 125-labeled IL-18 to the IL-18 receptor was blocked by neutralizing anti-hul L-18 monoclonal antibodies. In this manner, iodination did not affect the receptor binding of L-18 and did not mask the neuiralizing epitopes in IL-1 8. I L-18 125 l was used to de-neutralize the mode of neu- ralization and potency of anti-IL antibodies. -1 8 in the Receptor Union Test.
EXAMPLE 2: Generation and isolation of anti-IL-18 antibodies Example 2.1: Assays to identify anti-IL-18 anti-cusnes. Throughout Example 3 the following assays were used to identify and characterize anti-IL-18 antibodies unless is established in another way.
Example 2.1. A: ELISA An ELISA was developed to screen for antibodies that bind human IL-1. In this ELISA, biotinylated NEM-hul L-1 8 (see Example 1 .2.B) was captured either by goat ampi-bioinylated IgG or in streptavidin-coated plates. Hybridoma or B-cell supernatans were applied and the antibodies bound by IL-1 8 were determined using HPG-conjugated anti-human IgGs, following the standard ELISA standard probes well known in the maige.
Example 2.1.B: Affinity determinations using BIACORE technology The BIACORE assay (Biacore, Inc., Piscataway, NJ) determines the affinity of antibodies with kinetic measurements of constants of inactive, aciva scale. The antibodies are capitulated on a biosensor chip by means of a covalently linked secondary antibody (eg, goat anti-human IgG or anti-mouse IgG) and then Variani concentrations of recombinant L-18 are applied. The union is recorded as a function of time and consianids of a cinefic scale are calculated. In this essay, the acíiva scales with fast as 1 06M "1s" 1 and the inactive scales with lenías as 1 0"6s" 1, can be measured.
Example 2.1.C: Epitope Mapping BIACORE technology was used to map the epitopes recognized by antagonisms I L-8 as well as ani-I antibodies L-18. In summary, an anolygonism I L-1 8 was capitated on the BIACORE chip and rhu-I L-1 8 bound to the immobilized reagent. The union of the other ani-angonysia ani-IL-1 8 to this compiejo was thus tested. The simultaneous union of two reagents shows that both recognize distinct epííopos.
Example 2.2. : Generation of anti-IL-18 HuMAbs Using XENOMOUSE The XENOMOUSE transgenic mouse technology (Abgenix, I nc., Fremoní, CA) was used to obtain human anti-human 11-1 8 monoclonal antibodies (HuMAbs). This technology consists of transgenic mice carrying human variable heavy adena site carrying VH, DH and JM, cmu, Cdelia and a heavy chain site consisting of unique human IgG and light chain gene sites. In immunization with an antigen of interest, these mice generate human complement antibodies for the animal.
Example 2.2. A: Immunization of XENOMOUSE with IL-18 antigen The XENOMOUSE animals were immunized by means of a pads pad for all injections. The total volume of each injection was 50 μl per root, 25 ul per pad of paia. The initial immunization injection contained 40 ug of human 1L-18 (NEM-rhu-IL-1 8 in pyrogen-free DPBS mixed 1: 1 v / v with Gold TiirMax per root.) Subsequent groups were made with 40 ug of IL -18 human in pyrogen-free DPBS mixed with 25 ug of Adju-Phos (aluminum phosphate gel) per root for six times, then a final group of 40 ug of human IL-18 in pyrogen-free DBPS without adjuvant per root The animals were immunized on days 0, 4, 8, 11, 17, 21, 25 and 35 for this proiocolo.The fusions were carried out on day 39. Following the immunization regime described above, the roots were euhenized, then Lumbar and inguinal lymph nodes were recovered.
Example 2.2. B: Generation of Hybridoma The lymphocytes were released by mechanical rupture of lumbar and inguinal lymph nodes, obtained according to Example 2.2. A, using an iodine tri-urator, and suppressed T cells by CD90 negative selection. Hybridoma fusion was performed by washing the washed enriched B cells and P3X63Ag.86.53 non-secretory myeloma cells acquired from ATTC, # caí. CRL 1 580 (Kearney et al., J. Immunol., 123, 1 979, 1548-1550) in a ratio of 1: 1. The cell mixture was loosely formed by cenrifugation at 80 g. After the complete removal of the supernatant, the cells were drained with 2-4 mL of Pronasa solution (CalBiochem, San diego, CA, # cat.53702, 0.5 mg / ml in PBS) for no more than 2 minutuses. Then 3-5 ml of FBS was added to degrade the enzyme activity and the suspension was adjusted to 40 ml of volume by using the electroless cell fusion solution, ECFS (0.3 M Sucrose, Sigma-Aldrich, Yes. Louis, MO; # Ca.S7903, 0.1 mM Magnesium Acetone, Sigma, # Cat.M2545, 0. 1 mM Calcium Acetate, Sigma-Aldrich, St. Louis, MO, # Cat C4705). The supernatant was removed after sterilization and the cells were resuspended in 40 ml of ECFS. The wash cycle was re-coated and the fresh cells were resuspended in ECFS at a concentration of 2 × 10 6 cells / ml. The electro-cellular fusion was performed using a fusion generator, model ECM2001, Genetronic, Inc., San Diego, CA. The size of the fusion chamber used was 2.0 ml using the following instruments: condition of alignment: volyaje: 50 v, time: 50 s; Membrane rupture at: volyaje: 3000 v, time: 30 μs; Post-fusion clamping time: 3s. After fusion, the cells were resuspended in the hybridoma fusion medium: DMEM (JRH Biosciences), 15% FBS (Hyclone), containing 0.5XHA (Sigma-Aldrich, St. Louis, MO; #cat A9666), and was supplemented with L-glutamine, pen / syrip, OPI (oxaloaceia, pyruvate, bovine insulin) (iodine from Sigma) and I L-6 (Boehringer, Mannheim, Indianapolis, iN) for culture at 37 ° C and 1 0% CO2 in air. The cells were laminated in 96-well tissue culture plates with a flat bottom at 4x1 04 cells per well. Cultures were maintained in hybridoma fusion medium for 2 weeks before being transferred to the Hybridioma medium: DMEM (JRH Biosciences, Lenexa, KS), 15% FBS (Hyclone, Logan, UTA), and supplemented with L-glutamine, pen. / strip, OPI (oxolactam, pyruvate, bovine insulin) (all from Sigma) and IL-6 (Boehringer Mannheim, Indianapolis, IN). Hybridomas were selected for super survival on 0.5 XHA of hybridoma fusion media and supernatants from those cavities containing hybridomas were selected for antigen reactivity by ELISA. The ELISA format included incubating the supernatants on antigen-coated plates (plates coated with human L-18) and detecting human amphi- human IL-1 binding antibodies using horseradish peroxidase (HRP), human anti-human IgG. labeled mouse, then all positive samples were confirmed by two parallel ELISA grypos, which included incubating the supernadani on antigen-coated plates (plates coated with human IL-1) and detecting human anti-human IL-18 binding antibodies using horseradish peroxidase (HRP), Kappa chain and gamma-labeled anti-human gamma. Cloning was performed in selected positive antigen cavities using limited dilution laminate. Plates were visually inspected for the presence of single colony growth and supernatants from single colony cavities were then selected by antigen-specific ELISA as described above. The highly reactive clones were tested to verify the purity of the human kappa and gamma chain by ELISA using a Luminex instrument.
Table 5 Neutralization potency of Anti-IL-18 HuMAbs in KG-1 bioassay In short, the PCR primers for heavy chain variable regions were designed in such a way that they could be inserted into a pEF-BOS plasmid of casein which contains an IgG signal peptide and human lgG1 constant region sequence [wild type (SEQ ID NO: 2) or inactive mutant (SEQ ID NO: 3)]. The forward VH PCR primer contained Nrul restriction site, as did the nucleotide sequence of the signal peptide. The reverse VH PCR primer contained the Sali reslice site which was also made at the 5 'end of the Fe gamma-1 sequence. The VH PCR fragments were digested with Nrul / Sall and cloned into human IgG1 pEF-bos or human IgG1 mutant constructs pEF-BOS. The full light chain genes were moved in pEF-BOS vector in their existing Kappa format of pCADN vectors by Hindlll restriction digestion, filling the overhangs with T4 polymerase, followed by Notl digestion. These fragments of bluní / Noil light chain were cloned in this manner, neighbor pEF-BOS digested by Srff / Notl. The VH and VL regions of anties were cloned from original hybridoma strains. RNA was prepared from anty producing cells, RT-PCR was performed with the primers designed as described above, ie, Nrul / Sall primers for primers VH and Nrul / BsiWI pair VL. The Kappa and IgG 1 chains of full length were placed in cassette vectors. The selected anties were further modified. Anties that occur naturally have either glu- amine (O) or glucamay (E) as the NH2 terminus of light and / or heavy chain. The production of anties with Q as the iodine NH2 produces the heyerogenidad of iodine NH2 due to the cyclization of the residue of gluíamina to a glufamafo. Therefore, the glutamine residue in the NH2 terminal of some of the anties was mutated for glutamate. Also two residues, Leucine234 and Leucine235 in the obsessed region of Fe, were mutated to prevent the effector functions of the anty. In summary, Leucine 234 and Leucine 235 each were replaced by a residue of Alanine using standard molecular biological techniques (Lund, J. et al., J. Immunology (1991) 147: 2657-2662; Winner, et al., PAT US 5,648,260; 5624821; 5, 5,624,821). These mutated anties of Fe were named (mg 1). These mutants were also characterized in Examples 2.2.J.6, below.
Example 2.2. F: Characterization of selected neutralizing anti-IL-18 anties Several anti-human recombinant IL-18 anties with germline sequences were produced in mammalian cells, purified and functionally characterized in several trials (see Table 6).
Table 6: In vitro antigen binding, cell assay and column sequence characteristic of some anti-IL-18 anties KG-1 (ATCC # CCL-246) is a human myelomonocytic cell line expressing constiitutively low levels of functional L-18 receptor. Travazing with TNF regulates the IL-18Ra and ß subunits of the functional I L-18 receptor in these cells. The KG-1 bioassay was performed by incubating KG-1 cells treated by TFN with recombinant human IL-18 (rhu-IL-1 8) and determining the level of IFNα production. Human induced IL-18 by an ELISA (Konishi, K., et al (1997) J. Immunol. Methods 209: 187-191). The KG-1 bioassay was used to determine the potency of oligonazation 1-18. For example, ani-IL-1 anties were incubated with different concentrations of rhu-IL-18 and incubated with KG-1 cells brought by TNF in a 96-well plate for 16-1 8 hours at 37 ° C. The supernatants were tested and collected for levels of IFN? human by an ELISA. This assay can measure IC50 values down to 4x1 0"1 1 6 x1 0" 11M of an I L-18 antagonist. The resulting plasmid-containing sequences (exclusive of pUC1 9) for the following genes or regulatory elements in the following order: 5'-CMV enhancerUltimate promoter, major adenovirus, human immunoglobulin signal peptide, heavy chain immunoglobulin variable region 2.5 (E) mg 1, human gamma-1 immunoglobulin constant region, SV40 polyadenylation sequence, human gastrin transcription termination sequence, SV40 duplication origin (SV40 promoter / enhancer), murine dihydrofolate reducidase sequence, Herpes Simplex virus thymidine kinase polyadenylation sequence, CMV enhancer, last major adenovirus promoter, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa constant region, and SV40 polyadenylation sequence 3 '. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The vecfor also codified the expression of the mouse DHFR gene, which allowed the selection of cells transformed by virfud from their ability to grow in culíivo in the absence of nucleóíidos.
Example 2.2.G 2: Transfection of Expression Vector in Major Cell Strain Cell stock, CHO DUX B 1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1980) ), defective in the expression of the dihydrofolate redctase gene (DHFR), was used for transfection of the expression partner described in Example 2.2.G 1. CHO DUX B1 1 cells were transfected with the vector using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology; Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman , JG, Smith, JA, and K. Struhl eds; Wiley Iníerscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The plates were incubated at 37 ° C for two hours. One hundred fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of regulated saline 2 x Hepes (HeBS) in a 50 ml conical tube. KG-1 (ATCC # CCL-246) is a human myelomonocytic cell line that constitutively expresses low levels of functional L-18 receptor. Training with TNF regulates the subunits I L-1 8Ra and ß of the functional IL-1 8 receptor in these cells. The KG-1 bioassay was performed by incubating KG-1 cells irradiated by TFN with recombinant human L-18 (rhu-IL-1 8) and depleting the level of IFN? Human induced by IL-1 8 by an ELISA (Konishi, K., et al (1 997) J. Immunol. Methods 209: 1 87-191). The KG-1 bioassay was used to defer the nickiliary fractionation potency of anisogonysias 1-1 8. For example, the AI-1 L-1 8 antibodies were incubated with different concentrations of rhu-IL-1 8 and thus incubated with KG cells. -1 brought by TNF in a 96-well plate for 16-1 8 hours at 37 ° C. The supernatans were tested and collected for levels of I FN? human by an ELISA. This test can measure the IC50 values down to 4x10"1 1 6 x10" 1 1 M of an anonymysia I L-1 8.
The mixture was swirled for 5 seconds and incubated at ambient temperature for 20 minutes. One ml was dissipated almost on each plate (still in F12) and the plates were incubated at 37 ° C for 4 hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock continued for a minute after which DMSO was diluted by the addition of 5 ml of phosphate-buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight. Thereafter, the medium was changed to Gibco alpha MEM without nucleosides with 5% dialyzed fetal bovine serum (FBS), and six hours later the cells were culíivaron in plates of 96 cavities as follows. Cells from the 10 cm plates were harvested using trypsin digestion and resuspended in a total of 300 ml of Gibco alpha MEM without nucleosides with 5% serum. Twenty, 96-well plates were grown in 10 ml / plate, 1 00 1 / well. One hundred ml of the same medium was added to 100 ml of remaining cells and 20 plates of 96 additional cavities were cultured as above. (Esío was a second dilution). The medium was changed in the 96-well plates one week later and again a week after the procedure. The non-nucleoside MEM alpha medium was used to select cells expressing DHFR and, therefore, the expression vector.
Example 2.2.G 3: Selection of producer cells 2.5 (E) mg1 The culture supernatants of transfected CHO cells were tested for the presence of secreted antibody 2.5 (E) mg 1 using an ELISA specific for human IgG. Once a group of CHO transferases has been selected for the expression of a human antibody, an additional selection was used to isolate those cells that have amplified the number of copies of the expression vector inged in the CHO genome. Drug meíotrexate (MTX) was used for the selection of amplified strains. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were named after another MTX selection cycle of higher concentration., and were tested for the production of immunoglobulin. 2.5 (E) mg 1 expressing CHO cells were cultured in a bioreact of 1 to 1 5 cells and the antibody production was determined to be -1.0 g / L in a two week run.
Example 2.2.H: Physicochemical Characterization of 2.5 (E) mg1 derived from CHO cell The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg 1, was performed. The experimentally determined molecular weight of 2.5 (E) mg 1 was approximately 149 kDa, in good agreement with the theoretical molecular weight. Using Peptide mapping techniques (K Biemann Annu, Rev. Biochem, 1992 61 977-1010, D A. Lewis Accelerated Articles, Anal, Chem. 1994, 66, 585-595) it was confirmed that 2.5 (E) mg 1 had the correct N terminal tanio for light and heavy chains. There was very little heavy chain C-terminal variability, as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy terminus. Each heavy chain 2.5 (E) mg 1 contained a single N-linked glycosylation site with oligomannose and complex, fucosylated bianienar structures with 0, 1 and 2 residues of iodine galacose.
Example 2.2.1: Solubility and Stability of 2.5 (E) mg1 CHO cell derivative 2.5 (E) mg 1 purified was soluble for at least 62 mg / mL at pH 5, 6 and 7 regulators for a minimum of 4 weeks. The studies of accelerated esíabilidad with 2.5 (E) mg 1 to 37 ° C in regulating slopes were made to identify the tests of stability indication and the pH of optimal long-term storage. Samples were pooled in weekly samples for size exclusion HPLC analysis and SDS-PAGE to be tested for aggregation and fragmen- tation, LC-MS / MS peptide mapping for S-S linked deification, ani- native-ELISA and / or bioassay based on cell for measurement of activity, and HPLC of cation iníercambio and iso-Asp quantification for measurement of charge heyerogeneity. The preliminary analysis of the samples by SEC (size exclusion chroma- ography), SDS-PAGE and cation chromatography showed that all three tests indicated stability and, therefore, 2.5 (E) mg 1 is more stable at ~ pH6. The cells of the 1 cm plates were harvested using the digestion of ipsin and resuspended in a 300 ml iodine of Gibco alpha MEM without nucleosides with 5% serum. Veinie, 96-well plates were grown in 10 ml / plate, 100 1 / well. One hundred ml of the same medium was added to 100 ml of Residen cells and 20 plates of 96 additional cavities were cultured as above. (Esío was a second dilution). The medium was changed in the 96-well plates a week later and again a week after this. The MEM alpha medium without nucleosides was used to select cells expressing DHFR and therefore the expression partner.
Table 7 Biacore analysis of cytosine binding by 2.5 (E) mg1 The plasmid sequences contained proveíes (exclusive of pUC19) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, promoted last major adenovirus, human immunoglobulin signal peptide, variable region of heavy chain immunoglobulin 2.5 ( E) mg 1, region consisting of human gamma-1 immunoglobulin, SV40 polyadenylation sequence, human gasyrin transcription elimination sequence, SV40 duplication origin (promoter / SV40 enhancer), murine dihydrofolate reductase sequence, polyadenylation sequence of Herpes Simplex virus mimidine kinase, CMV enhancer, last major adenovirus promoter, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa constant region, and SV40 polyadenylation sequence 3 '. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The neighbor also encoded the expression of the root DHFR gene, which allowed the selection of cells transformed by viriud from their ability to grow in culture in the absence of nucleoids.
Example 2.2.G 2: Transfection of Expression Vector in Major Cell Strain The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1 980) ), deficient in the expression of the dihydrofolate redcíasa gene (DHFR), was used for transfection of the expression partner described in Example 2.2. G 1 CHO DUX B1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the maize (Currení Proíocols in Molecular Biology; Ausubel, F.V., Brení, R., Moore, D.M. , Kingston, R.E., Seidman, J.G. , Smith, J.A. , and K. Struhl eds; Wiley Iníerscience, N .Y. , N.Y. (1 990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The plates were incubated at 37 ° C for two hours. One hundred fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of regulated saline 2 x Hepes (HeBS) in a 50 ml conical tube.
Sequences contained plasmid turnout (exclusive of pUC19) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, adenovirus major ulterior promoter, human immunoglobulin signal peptide, heavy chain immunoglobulin variable region 2.5 ( E) mg1, human gamma-1 immunoglobulin constant region, SV40 polyadenylation sequence, human gasyrin transcription termination sequence, duplication SV40 origin (promoior / SV40 enhancer), murine dihydrofolate reductase sequence, kinase polyadenylation sequence Herpes simplex virus mimidine, CMV enhancer, promoter of the major adenovirus, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa region, and SV40 polyadenylation 3 'sequence. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The neighbor also encoded the expression of the mouse DHFR gene, which allowed the selection of cells transformed by viriud from their ability to grow in culture in the absence of nucleoids. The mixture was swirled for 5 seconds and incubated at ambient temperature for 20 minutes. One ml was dissipated almost on each plate (still in F12) and the plates were incubated at 37 ° C for 4 hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The shock of DMSO continued for a minute after which DMSO was diluted by the addition of 5 ml of phosphate-buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight. The next day, the medium was changed to Gibco alpha MEM without nucleosides with 5% dialyzed fetal bovine serum (FBS), and six hours later the cells were cultured in 96-well plates as follows. Cells from the 10 cm plates were harvested using trypsin digestion and resuspended in a 300 ml bead of Gibco alfa MEM without nucleosides with 5% serum. Twenty, 96-well plates were grown in 10 ml / dish, 100 1 / well. One hundred ml of the same medium was added to 100 ml of Restani cells and 20 additional 96-well plates were cultured as above. (This was a second dilution). The medium was changed in the 96-well plates a week later and again a week after this. The MEM alpha medium without nucleosides was used to select cells expressing DHFR and therefore the expression vector.
Example 2.2.G 3: Selection of producer cells 2.5 (E mg1) Culture supernatants from transfected CHO cells were tested for the presence of secreted antibody 2.5 (E) mg 1 using a specific ELISA for human IgG. Transfection of CHO has been selected for the expression of a human antibody, a further selection was used to isolate those cells that have amplified the copy number of the expression vector integrated in the CHO genome.The drug methotrexate (MTX) was used to The selection of amplified strains The cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin.The MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken out of another MTX selection cycle. of higher concentration, and were tested for immunoglobulin production 2.5 (E) mg 1 expressing CHO cells was cultured were in a bioreact of 1 to 15 liters and antibody production was determined to be -1.0 g / L in a two week run.
Example 2.2.H: Physicochemical Characterization of 2.5 (E) mg1 derived from CHO cell The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg 1, was performed. The experimentally determined molecular weight of 2.5 (E) mg1 was approximately 149 kDa, in good agreement with the molecular weight. Using techniques of Péplido mapping (K Biemann Annu, Rev. Biochem. 1992 61 977-1010; D A. Lewis Accelerated Articles, Anal. Chem. 1994, 66, 585-595) it was confirmed that 2.5 (E) mg1 had the correct N-terminal for both light and heavy chains. There was very little heavy chain C-terminal variability, as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy ferminal. Each heavy chain 2.5 (E) mg1 contained a single N-linked glycosylation site with oligomannose and complex, bucosenar fucosylated structures with 0, 1 and 2 residues of iodine galacose.
Example 2.2.1: Solubility and Stability of 2.5 (E) mg1 CHO cell derivative 2.5 (E) mg 1 purified was soluble for at least 62 mg / mL at pH 5, 6 and 7 regulators for a minimum of 4 weeks. The accelerated esivity studies with 2.5 (E) mg1 at 37 ° C in regulatory stages were performed to identify the stability indication tests and the optimal long-term storage pH. The preliminary chemical and physical characterization of CHO derived 2. 5 (E) mg 1, was performed. The experimentally demineralized molecular weight of 2.5 (E) mg1 was approximately 149 kDa, in good agreement with the molecular weight. Using Peptide mapping techniques (K Biemann Annu, Rev. Biochem, 1992 61 977-1 01 0, D A.
Lewis Accelerated Aríicles, Anal. Chem. 1994, 66, 585-595) it was confirmed that 2.5 (E) mg 1 was the correct N-terminal for both light and heavy chains. There was very little heavy chain C-terminal variability, such as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy terminus.
Each heavy chain 2.5 (E) mg 1 contained a unique N-linked glycosylation site with oligomannose and complex, fucosylated biatenary structures with 0, 1 and 2 terminal galactose residues.
Samples were taken at weekly intervals for analysis by Size exclusion HPLC and SDS-PAGE to be tested for aggregation and fragmentation, LC-MS / MS peptide mapping for SS linked deification, antigen-ELISA and / or cell-based bioassay for activity measurement, and HPLC exchange of. cation and iso-Asp quantification for measurement of charge heterogeneity. The preliminary analysis of the samples by SEC (size exclusion chromatography), SDS-PAGE and cation exchange chromatography showed that all three tests indicated stability and therefore 2.5 (E) mg 1 is more stable at ~ pH6.
Table 8 IL-18 Bonding Properties of 2.5 (E) mg1 and Reagents Reference Table 8 Neutralization Potencies of 2.5 (E) mg1 and Reagents Reference -jO The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg 1, was performed. The experimentally deferred molecular weight of 2.5 (E) mg1 was approximately 149 kDa, in good agreement with the molecular weight. Using Peripid mapping techniques (K Biemann Annu, Rev. Biochem, 1992 61 977-1010, D A. • 15 Lewis Acceleraíed Arlicles, Anal. Chem. 1994, 66, 585-595) it was confirmed that 2.5 (E) mg 1 had the correct N-terminal tanium for light and heavy chains. There was very little heavy chain C-terminal variability, as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy terminus.
Each heavy chain 2.5 (E) mg 1 contained a single N-linked glycosylation site with oligomannose and complex, bucosenar fucosylated structures with 0, 1 and 2 terminal galacose residues.
Table 9A In Vitro Efficacy of 2.5 (E) mg 1 administered i.p. in HuPBMC-SCID mouse model Table 9B In Vitro Efficacy of 2.5 (E) mg 1 administered i.v. in HuPBMC-SCID mouse model The resulting plasmid sequences result (exclusive of pUC19) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, major promoter, adenovirus, human immunoglobulin signal peptide, heavy chain immunoglobulin variable region 2.5 (E) mg 1, region consisting of human gamma-1 immunoglobulin, SV40 polyadenylation sequence, human gasyrin transcription cross-section, SV40 duplication origin (promoter / SV40 enhancer), murine dihydrofolate reductase sequence, polyadenylation of Herpes Simplex virus thymidine kinase, CMV enhancer, promoter of the major adenovirus, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa region, and SV40 polyadenylation sequence 3 '. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The neighbor also encoded the expression of the root DHFR gene, which allowed the selection of transformed cells by virtue of their ability to grow in culture in the absence of nucleotides.
Example 2.2.G 2: Transfection of Expression Vector in Major Cell Strain Cell stock, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1980)) , defective in the expression of the dihydrofolate redctase gene (DHFR), was used for transfection of the expression vector described in Example 2.2.G 1. CHO DUX B 1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman, JG, Smith, JA, and K. Struhl eds; Wiley Iníerscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plate. The plates were incubated at 37 ° C for two hours. Half a hundred micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of 2 x Hepes buffered saline (HeBS) in a 50 ml conical tube. The resulting mixture was swirled for 5 seconds and incubated at room temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated 37 ° C for four hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight. The next day, the medium was changed to Gibco alpha MEM without nucleosides with 5% dialyzed fetal bovine serum (FBS), and six hours later the cells were cultured in 96-well plates as follows. Cells from the 10 cm plates were harvested using trypsin digestion and resuspended in a total of 300 ml of Gibco alpha MEM without nucleosides with 5% serum. Twenty, 96-well plates were grown in 10 ml / plate, 100 1 / well. One hundred ml of the same medium was added to 100 ml of Trannian cells and 20 plates of 96 additional cavities were culíivaron according to anleriormenie. (This was a second dilution). The medium was changed in the 96-well plates a week later and again a week after this. The MEM alpha medium without nucleosides was used to select cells expressing DHFR and therefore the expression vector.
Example 2.2.G 3: Selection of producer cells 2.5 (E) mg1 Culture supernatants of transfected CHO cells were tested for the presence of secreted antibody 2. 5 (E) mg 1 using a specific ELISA for human IgG. Once a group of CHO transfectants has been selected for the expression of a human antibody, an additional selection was used to isolate those cells that have amplified the copy number of the expression vector inged in the CHO genome. Drug meíotrexate (MTX) was used for the selection of amplified strains. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati, Acad SCI USA 77: 4216-4220 (1 980)), defective in the expression of the dihydrofolate redctase gene (DHFR), it was used for transfection of the expression vector described in Example 2.2. G 1 CHO DUX B1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman , JG, Smith, JA, and K. Struhl eds; Wiley Iníerscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to the MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken through another cycle of selection in MTX concentration higher, and were tested for the production of immunoglobulin. 2.5 (E) mg 1 expressing CHO cells were cultured in a bioreact of 1 to 15 liíros and antibody production was determined to be -1 .0 g / L in a two week run.
Example 2.2.H: Physicochemical Characterization of 2.5 (E) mg1 derived from CHO cell The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg 1, was performed. The experimentally determined molecular weight of 2.5 (E) mg1 was approximately 149 kDa, in good agreement with the theoretical molecular weight. Using Peptide mapping techniques (K Biemann Annu, Rev. Biochem, 1992 61 977-1 01 0, D A. Lewis Accelerated Aricles, Anal, Chem. 1 994, 66, 585-595) it was confirmed that 2.5 (E) mg 1 I get the correct N terminal for both light and heavy chains. The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redctase gene (DHFR), was used for transfection of the expression described in Example 2.2.G 1. CHO DUX B1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman , J .G., Smith, JA, and K. Struhl eds; Wiley I nterscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mix was added one drop at a time. There was very little heavy chain C-terminal variability, as 99% of molecules 2.5 (E) mg 1 lacked terminal lysine of heavy chain carboxy. Each heavy chain 2.5 (E) mg 1 contained a single N-linked glycosylation site with oligomannose and complex, bucosenar fucosylated structures with 0, 1 and 2 terminal galactose residues.
Example 2.2.1: Solubility and Stability of 2.5 (E) mq1 Derivative of CHO cell 2.5 (E) mg 1 purified was soluble for at least 62 mg / mL at pH 5, 6 and 7 regulators for a minimum of 4 weeks. Accelerated stability studies with 2.5 (E) mg 1 at 37 ° C in these regulators were performed to identify the stability indication assays and the optimal long-term storage pH. Samples were taken at weekly intervals for size exclusion HPLC analysis and SDS-PAGE to be tested for aggregation and fragmentation, LC-MS / MS peptide mapping for SS-linked detection, ELISA-antigen and / or cell-based bioassay for measurement of activity, and HPLC of cation iníercambio and iso-Asp quantification for measurement of load heyerogeneity. The preliminary analysis of the samples by SEC (size exclusion chroma- ography), SDS-PAGE and chromatography of the cation in vitro showed that all the tests indicated stability and, therefore, 2.5 (E) mg 1 is more stable to ~ pH6.
Table 10: Residues of mutations L234 and L235 do not affect the affinity or potency of neutralization to Alanine of 2.5 (E) mg1 THE. Proc. Nati Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redctase gene (DHFR), was used for transfection of the expression vector described in Example 2.2. G 1 CHO DUX B1 1 cells are transfected with the vector using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology; Ausubel, FV, Brent, R., Moore, DM, Kingsion, RE, Seidman , JG, Smiíh, JA, and K. Struhl eds; Wiley Iníerscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours. One hundred fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microlives of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to the mixture.
Table 1 1 Demonstration of the faults of 2.5 (E) mg1, in contrast to 2.5 (E) wtg1, to FcyRI binding on U937 cells (data shown as MF1 +/- SD) Table 12 Binding of 2.5 (E) wtg1 and 2.5 (E) mg1 to FCyR II on K562 cells after traversing the link (data shown as MFI +/- SD) Table 13. Demonstration of the failure of 2.5 (E) mg1, in contrast to 2.5 (E) wtg1, for binding to C1 q by ELISA (data shown as OD405 +/- SD) Table 14 Summary of key pharmacokinetic parameters of 2.5 (E) mg1 derived from a single intravenous dose in mice The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati, Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redcíasa gene (DHFR), Use for the transfection of the expression partner described in Example 2.2.G 1.
CHO DUX B1 1 cells are transfected with the vector using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman , JG, Smith, J .A., And K. Struhl eds; Wiley Interscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The plates were incubated at 37 ° C for two hours. One hundred fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microlipres of 2.5M CaCl2 was added and the DNA mixture was added one go at a time to the mixture.
Table 15 93-10C inhibits in vivo production of LPS-induced IFNg Group mulFNg (pg / ml)% In h ibition Sequences contained plasmid turnout (exclusive of pUC1 9) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, major promoter, adenovirus, human immunoglobulin signal peptide, heavy chain immunoglobulin variable region 2.5 (E) mg1, region consisting of human gamma-1 immunoglobulin, SV40 polyadenylation sequence, human gasrrin transcription elimination sequence, SV40 duplication origin (promoter / SV40 enhancer), murine dihydrofolate reductase sequence, polyadenylation sequence of Herpes Simplex virus thymidine kinase, CMV enhancer, last major adenovirus promoter, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa constant region, and SV40 polyadenylation sequence 3 '. The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin THE. Proc. Nati Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redcíasa gene (DHFR), was used for expression of the expression partner described in Example 2.2.G 1. CHO DUX B1 1 cells were transfected with the vector using the well-known calcium phosphate precipitation method in the maize (Current Protocols in Molecular Biology; Ausubel, FV, Breni, R., Moore, DM, Kingston, RE, Seidman , JG, Smith, JA, and K. Struhl eds; Wiley Interscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and a mixture of DNA was added one drop at a time to the mixture. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The vector also encoded the expression of the mouse DHFR gene, which allowed the selection of transformed cells by virtue of their ability to grow in culture in the absence of nucleotides.
Example 6: Transfection of Expression Vector in Major Cell Strain Cell stock, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redctase gene (DHFR) was used for transfection of the expression vector described in Example 2.2.G 1. CHO DUX B1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology; Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman, JG, Smith, JA, and K.
Struhl eds; Wiley Interscience, N .Y. , N .Y. (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin LA Proc. Nati. Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redcíasa gene (DHFR), was used for transfection of the expression partner described in FIG. Example 2.2.G 1. CHO DUX B 1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman, JG, Smith, JA, and K. Struhl eds; Wiley lnterscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each plaque. The plates were incubated at 37 ° C for two hours. One hundred fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three microliths of 2.5M of CaCl2 was added and a mixture of DNA was added one go at a time to the mixture. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of 2 x Hepes buffered saline (HeBS) in a 50 ml conical tube. The resulting mixture was swirled for 5 seconds and incubated at ambient temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated at 37 ° C for four hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight. The next day, the medium was changed to Gibco alpha MEM without nucleosides with 5% dialyzed fetal bovine serum (FBS), and six hours later the cells were cultured in 96-well plates as follows. Cells from the 10 cm plates were harvested using trypsin digestion and resuspended in a 300 ml bead of Gibco alfa MEM without nucleosides with 5% serum. Twenty, 96-well plates were grown in 10 ml / dish, 100 1 / well. One hundred ml of the same medium was added to 100 ml of Restani cells and 20 additional 96-well plates were cultured as above. (This was a second dilution). The medium was changed in the 96-well plates a week later and again a week after this. The MEM alpha medium without nucleosides was used to select cells expressing DHFR and therefore the expression partner.
Example 2.2.G 3: Selection of producer cells 2.5 (E) mg1 Culture supernatants from transfected CHO cells were tested for the presence of secreted antibody 2.5 (E) mg 1 using an ELISA specific for human IgG. Once a group of CHO transfectants has been selected for the expression of a human antibody, further selection was used to isolate those cells that have amplified the copy number of the expression vector integrated into the CHO genome. The drug methorerexate (MTX) was used for the selection of amplified strains. The culinos that grew in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken through another selection cycle in MTX of higher concentration, and were tested for immunoglobulin production. 2.5 (E) mg 1 expressing CHO cells were cultured in a bioreact of 1 to 15 liíros and the production of an antibody was determined to be -1.0 g / L in a two week run.
Example 2.2.H: Physico-chemical characterization of 2.5 (E) mg1 derived from CHO cell The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg1, was performed. The experimentally determined molecular weight of 2.5 (E) mg 1 was approximately 149 kDa, in good agreement with the theoretical molecular weight. Using techniques of Pepiid mapping (K Biemann Annu, Rev. Biochem, 1992 61 977-1010, D A. Lewis Accelerated Aricles, Anal, Chem. 1 994, 66, 585-595) it was confirmed that 2.5 (E) mg1 was The N-terminal is used for light and heavy chains. There was very little heavy chain C-terminal variability, as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy-terminal. Each heavy chain 2.5 (E) mg1 contained a single N-linked glycosylation site with oligomannose and complex, fucosylated baryary structures with 0, 1, and 2 residues of the iodine galacose. The drug metabolism (MTX) was used for the selection of amplified strains. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were identified from another MTX selection cycle of higher concentration, and were tested for immunoglobulin production.
Example 2.2.1: Solubility and Stability of 2.5 (E) mg1 CHO cell derivative 2.5 (E) mg 1 purified was soluble for at least 62 mg / m L at pH 5, 6 and 7 regulators for a minimum of 4 weeks. Accelerated cleavage studies with 2.5 (E) mg 1 at 37 ° C in regulatory environments were performed to identify the stability indication tests and the optimal long-term storage pH. Each heavy chain 2.5 (E) mg 1 contained a single N-linked glycosylation site with oligomannose and complex, fucosylated biatenary structures with 0, 1 and 2 residues of the iodine galacose. The drug metabolite (MTX) was used for the selection of amplified smears. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX-resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken through another selection cycle in MTX of higher concentration, and were tested for immunoglobulin production. Samples were taken in weekly samples for size exclusion HPLC analysis and SDS-PAGE to be tested for aggregation and fragmentation, LC-MS / MS peptide mapping for SS-linked detection, ELISA-antigen and / or cell-based bioassay for measurement of acidity, and HPLC of cation iníercambio and iso-Asp quantification for measurement of load heyerogeneity. The preliminary analysis of the samples by SEC (size exclusion chroma- ography), SDS-PAGE and in vitro cyaniography showed that all three tests indicated stability and therefore 2.5 (E) mg 1 is more stable to ~ pH6.
Table 16 In vivo suppression of the carriage-paw edema-inducing gene The plasmid sequences contained proveíes (exclusive of pUC19) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, major promoter, adenovirus, human immunoglobulin signal peptide, heavy chain immunoglobulin variable region 2.5 ( E) mg 1, region consisting of human gamma-1 immunoglobulin, SV40 polyadenylation sequence, human gastrin transcription termination sequence, duplication SV40 origin (SV40 promoter / enhancer), murine dihydrofolate reductase sequence, polyadenylation sequence of Herpes Simplex virus mimidine kinase, CMV enhancer, last major adenovirus promoter, human immunoglobulin signal peptide, 2.5. (E) light chain immunoglobulin variable region mgl, human immunoglobulin kappa constant region, and SV40 polyadenylation sequence 3 '. Each heavy chain 2.5 (E) mg 1 contained a single-linked glycosylation site with oligomannose and complex, fucosylated biannual esírucíuras with 0, 1 and 2 residues of ferminal galacíosa. Drug methotrexate (MTX) was used for the selection of amplified strains. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken through another MTX selection cycle of higher concentration, and were tested for immunoglobulin production. The coding regions were inserted downstream of strong viral promoters that led to the transcription of the antibody gene. The vector also encoded the expression of the mouse DHFR gene, which allowed the selection of transformed cells by virtue of their ability to grow in culture in the absence of nucleotides.
Example 2.2.G 2: Transfection of Expression Vector in Major Cell Strain The cell line, CHO DUX B1 1, (Uriaub, G. and Chasin THE. Proc. Nati Acad SCI USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate redctase gene (DHFR), was used for transfection of the expression vecfor described in Example 2.2.G 1. CHO DUX B1 1 cells are transfected with the neighbor using the calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology, Ausubel, FV, Brent, R., Moore, DM, Kingston, RE, Seidman , JG, Smith, J.A., and K. Struhl eds; Wiley Interscience, NY, NY (1990)) with the following modifications. The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours.
One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of 2 x Hepes buffered saline (HeBS) in a 50 ml conical tube. The resulting mixture was swirled for 5 seconds and incubated at room temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated at 37 ° C for four hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight. The next day, the medium was changed to Gibco alpha MEM without nucleosides with 5% dialyzed fetal bovine serum (FBS), and six hours later the cells were cultured in 96-well plates as follows. Cells from the 10 cm plates were harvested using trypsin digestion and resuspended in a total of 300 ml of Gibco alpha MEM without nucleosides with 5% serum. Twenty, 96-well plates were grown in 10 ml / dish, 100 1 / well. One hundred ml of the same medium was added to 100 ml of remaining cells and 20 additional 96-well plates were cultured as above. (This was a second dilution). The medium was changed in the 96-well plates a week later and again a week after this. The MEM alpha medium without nucleosides was used to select cells expressing DHFR and therefore the expression vector.
Example 2.2.G 3: Selection of producer cells 2.5 (E) mg1 The culture supernatants of transfected CHO cells were tested for the presence of secreted antibody 2.5 (E) mg 1 using an ELISA specific for human IgG. Once a group of CHO transfectants has been selected for the expression of a human antibody, further selection was used to isolate those cells that have amplified the copy number of the expression vector integrated into the CHO genome. The drug method (MTX) was used for the selection of amplified strains. Cultures grown in the presence of MTX were tested for their ability to produce immunoglobulin. The MTX resistant strains that expressed more antibody than the MTX-sensitive predecessors were taken through another MTX selection cycle of higher concentration and tested for immunoglobulin production. 2.5 (E) mg1 expressing CHO cells were cultured in a bioreact of 1 to 15 liíros and antibody production was determined to be -1.0 g / L in a two week run.
Example 2.2.H: Physicochemical Characterization of 2.5. { E) CH1 cell derivative mg1 The preliminary chemical and physical characterization of CHO derived 2.5 (E) mg 1, was performed. The experimentally determined molecular weight of 2.5 (E) mg 1 was approximately 149 kDa, in good agreement with the theoretical molecular weight. Using Peptide mapping techniques (K Biemann Annu, Rev. Biochem.1992 61 977-1010; D A. Lewis Accelerated Aricles, Anal. Chem. 1994, 66, 585-595) it was confirmed that 2.5 (E) mg 1 was the N-terminal for the light and heavy chains. There was very little heavy chain C-terminal variability, such as 99% of molecules 2.5 (E) mg 1 lacked lysine in the heavy chain carboxy terminus. Each heavy chain 2.5 (E) mg 1 contained a single N-linked glycosylation site with oligomannose and complex, fucosylated bianienar structures with 0, 1 and 2 residues of iodine galacose.
Example 3: Solubility and Stability of 2.5 (E) mg1 CHO cell derivative 2.5 (E) mg 1 purified was soluble for at least 62 mg / mL at pH 5, 6 and 7 regulators for a minimum of 4 weeks. Accelerated stability studies with 2.5 (E) mg1 at 37 ° C in these regulators were performed to identify the stability indication assays and the optimal long-term storage pH. Samples were taken at weekly intervals for size exclusion HPLC analysis and SDS-PAGE to be tested for aggregation and fragmentation, LC-MS / MS peptide mapping for SS-linked detection, ELISA-antigen and / or cell-based bioassay for measurement of activity, and HPLC of cation exchange and iso-Asp quantification for measurement of charge heterogeneity. The preliminary analysis of the samples by SEC (size exclusion chromatography), SDS-PAGE and cation exchange chromatography showed that all three tests indicated stability and therefore 2.5 (E) mg 1 is more stable at ~ pH6.
Table 17 Anti IL-18 mAb 93-10C delays the grouping and decreases the severity of CIA Treatment Meaning of the Assessment Arthritic The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of 2 x Hepes buffered saline (HeBS) in a 50 ml conical tube. The resulting mixture was swirled for 5 seconds and incubated at room temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated at 37 ° C for four hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight.
Tratamienío Meaning of Arthritic Assessment Irrigation period P < 0.05 vs. Rat IgG Example 7: Septic Arthritis I L-1 8 is an important factor in the pathogenesis of a mouse model of septic arthritis. This is not generally considered to be a model of RA, but it does share some inflammatory components and pathology related to RA. In this model, the disease is induced by injecting the live B streptococci (GBS) group into knee joints. The severity of the resulting arthritis correlates with local and systemic levels of IL-1β and IL-6, but not TNF (Tissi L, et al (1999) Infecí.Immunol.67: 4545-50). Levels I L-18 significance in the joints were detected as early as 12 hours post-injection with serotype IV (GBS) followed by production of maximum IL-18 after 5 days (-550 pg / ml in 1 C5 vs. IV). -30 pg / ml in IgG control). Elevated levels I L-1 8 were detected in the serum by day 5 of post injection (-1 80 pg / ml in 1 C5 brought vs. -20 pg / ml in conírol IgG). When 1 C5 was injected 1 hour after the administration of GBS, the inhibition in the frequency of arylic lesions from day 2 to day 10 was marked (arthritic index: 1.0 in 1 C5 trayed vs. 2.5 in conirol IgG). In addition, irradiation 1 C5 also resulted in the significant reduction in cytokine levels in joints that include IL-6 and I L-1β. (Data not revealed). . The plates were aspirated and 9 ml of F12 medium was added to each silver. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microliters of 2.5M CaCl2 was added and this DNA mixture was added one drop at a time to 3 ml of 2 x Hepes buffered saline (HeBS) in a 50 ml conical tube. The resulting mixture swirled for 5 seconds and was incubated at ambient temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated at 37 ° C for four hours. After the incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight.
Example 2.2.L.5: SLE The most studied models of lupus include strains of mice (MRL / Ipr and NZB / NZW F1) that spontaneously develop lipo lupus syndrome with severe glomerulonephritis, antibody production (anti-DNA, anti RNP etc. ), splenomegaly, lymphadenopathy, and a certain degree of arthritis and vasculiitis. Kidney inclusion is usually seen at 3-5 months of age, progresses rapidly, and for 6-10 months is fatal. Both mouse strains have been extensively studied to gain an understanding of clinical disease. The NZB / NZW F1 (B / W) mouse model (The Jackson Laboratory, Maine, USA) was selected as the most relevant model to evaluate the effects of exogenous I L-18 on lupus-like disease progression. The onset of disease progression in B / W mice is usually observed at 7-9 months of age and for 12-14 months it is mainly as a resulfate of renal failure. To investigate the role of IL-18 in lupus pathogenesis, B / W mice were brought daily with r-mulL-18 or vehicle control starting at 7 months of age. Kidney function was assessed by determining the degree of proteinuria. Daily treatment of B / W mice with 50 μg / kg of IL-18 led to accelerated onset of severe protelnuria as compared to the idraed group of PBS vehicle. The B / W rafons of I L-1 8 were also accelerated méridres mosíradas. These observations were consistent with those previously described for the MRL / lpr rafones and underline a pro-inflammatory role for I L-18 in autoimmune disease.
Table 18 Mice treated with anti-IL-18 Ab and unconscious mice with IL-18 develop less severe EAE disease Table 19 In vivo inhibition of ConA-induced liver inflammation 93-10C The plates were aspirated and 9 ml of F12 medium was added to each plate. The plates were incubated at 37 ° C for two hours. One hundred and fifty micrograms of DNA were dissolved in 2.7 ml of water in a 50 ml conical tube. Three hundred microlives of 2.5M CaCl2 was added and this DNA mixture was added one go at a time to 3 ml of 2 x Hepes regulated salt (HeBS) in a 50 ml conical tube. The resulting mixture was swirled for 5 seconds and incubated at room temperature for 20 minutes. One ml was distributed almost on each plate (still in F12) and the plates were incubated at 37 ° C for four hours. After incubation, the plates were aspirated and 2 ml of 10% DMSO in F12 was added to each plate. The DMSO shock was continued for one minute after which DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) for each plate. Plates were aspirated and washed twice in PBS. Ten ml of Gibco MEM alpha with nucleosides was added and the plates were incubated at 37 ° C overnight.
Table 21. Genes induced by IL18.
Table 22. Genes regulated by IL18 following the treatment of TNF. phospholipase .2 receptacle 1, U 1 7034 PLA2R1 IdOkD 29.6 28.7 lectin, galacioside binding, M5771 or soluble LGALS3, 3 (galectin 3) 27.5 25.4 penfaxin-related gene, X97748 PTX3 I L-1 induced 15.2 13.6 M27288 OSM oncosyin M 23.1 12.0 Lambda light chain X57809 variable region 1 0.9 1 0.0 interleukin 6 (interferon, 100081 IL6 beía 2) 9.2 9.4 D16583 HDC histidine decarboxylase 8.0 9.4 kallikrein 3, (specific prosyáíico X07730 KLK3 antigen) 5.6 8.8 HG31 1 1 - Clone Oven sapiens HH409 9.5 7.5 HT3287 unknown Hepatic nuclear factor M57732 TCF1 (FNH 1) 2.0 7.2 U77735 PIM2 pim-2 oncogen 7.1 7.1 U96094 SLN sarcolipin 12.2 6.1 phosphodiesierase 3B, cGMP-D50640 PDE3B inhibited 4.0 5.4 lysozyme (amyloidosis X14008 LIZ renal) 3.0 5.4 M91 036 HBG2 hemoglobin, gamma G 3.4 5.4 monoclin induced by X72755 MIÓ gamma interferon 5.2 5.2 Glutama receptor, AC000099 GRM8 meíaboírópico 8 2.3 4.3 D1 1428 PMp22 Proiein peripheral myelin 5.0 4.0 22 Table 23. Another potential IL1 S / IL12 selected transcription markers regulate four or more folds by IL18 + IL12 i reversible by 1252H in a human leukocyte sample determined using Affimetrix Genechips.
Table 25. Performance of interferon I in ten blood samples of human. P < 0.05 for inhibition of any antibody Table 26. MIG / CXCL9 performance in ten human blood samples. P < 0.05 for inhibition of any antibody Table 27. Performance MCP2 / CCL8 in ten human blood samples. p < 0.05 for inhibition of any antibody Table 28. Biacore analysis of cytosine bound by 2.13 (E) mg1 i 2.5 (E) mg1 Table 29. BIACORE analysis of antibody competition for 11-18 human binding The present invention is incorporated for reference in its well-known techniques in the field of molecular biology. These techniques include, but are not limited to, technical techniques in the following publications: Ausubel, F. M. ef al. eds. , Short Protocols in Molecular Biology (4th Ed. 1999) John Wiley & Sons, (ISBN 0-471 -32938-X). Lu and Weiner eds., Cloning and Expression Vecors for Gene Function Analysis (2001) BioTechniques Press. Westborough, MA. 298 pp. (ISBN 1 -881299-21 -X). and Dubel eds. , Antibody Engineering (2001) Springer-Verlag. New York 790 pp. (ISBN Older, RW &SB Primrose, Principles of Gene Manipulation: An Introduction to Generation Engineering (3d Ed. 1985) Blackwell Scientific Publications, Bosium, Siudies in Microbiology, V. 2: 409 pp. (ISBN 0-632-0131 8-4) Sambrook, J., Et al., Molecular Cloning: A Manual Laboraíory (2d Ed. 1989) Cold Spring Harbor Laboraíory Press, NY, Vols. 1 -3. (ISBN 0-87969-309-6 Winnacker, EL From Genes To Clones: Introduction to Gene Technology (1987) VCH Publishers, NY (translated by Horst 634 pp. (ISBN References US Patents 5,545, 806 5,545, 807 5,591, 669 5,612, 205 5,625, 126 5,625, 825 5,627, 052 5,633, 425 5,643, 763 5,661, 016 5,721, 367 5,770, 429 5,789, 215 5,789, 650 5,814, 318 5,912, 324 5,916, 771 5,939, 598 5,985, 6155,994, 6195,998, 2096,054, 4876,060, 2836,075, 6,091, 001 6,114, 598 6,130, 364 patent application publication 20030186374 US application. Serial No. 09 / 428,082 Other Patent Documents EP 712 931 EP 850 952 EP 864 585 EP 0 962 531 EP 600 JP 111,399194 IL 121554 AO WO 91/10741 WO 91/17271 WO 92/01047 WO 92/02551 WO 92/09690 WO WO 92 / 18619 WO WO 93/01288 WO 94/02602 WO 96/33735 WO 96/34096 WO WO 97/29131 WO WO WO O WO WO 99/25044 WO 99/37772 WO 99/37773 WO 99/45031 O WO 00/37504 WO WO 00/12555 WO 00/37504 WO 00/56772 WO O WO 02/72636.
Other References Adachi ef al. (1998) Immuniíy 9: 143-150 Akiía, K. et al., (1 997) J. Chem. 272,26595-26603 Azzazy H., and WE, (2002) 35: 425-445 Babcock, JS eí ( 1996) Proc. Nati Sci. USA 93: 7843-7848 Barbas ei (1 991) PNAS 88: 7978-7982 Bendele, A., et al. (1999) Toxicol Paihol. 27: 1 34-142 Bird eí (1988) Science 242: 423-426 Clackson ef al. (1991) Nature 352: 624-628 Dinarello, C. et al. (1 998) J. Leukoc. Biol. 63: 658-654 Dinarello, C. A. (1 999) Methods 19: 121-132 Dinarello, C. A. (1999) J. 103: 1 -24; Durocher et al. , Nucleic Acids Research 2002, Vol 30, No. 2 Fuchs et al. (1991): 1370-1372 ef al. (1991): 1373-1377 Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29: 128-145 Giege, R. and Ducruix, A. Barretí, Crystallization of Nucleic Acids and Proteins, a Practical Approach, 2nd ea., Pp. 20 1-16, Oxford University Press, New York, New York, (1999). Ghayur, T. ef al., (1997) Naíure 386: 619-623 Ghetie, V., eí al (1997) Nal. Bioiechnol. 15: 637-640 Gracie J.A., ef al., (2003) Journal of Leukocyte Biology 73.213-224 Gram et al. (1992) PNAS: 3576-3580. Green ef al. Nature Genetics 7: 13-21 (1994) Green and Jakobovits J. Exp. Med. 188: 483-495 (1998) Griffiths et al. (1 993) EMBO J 12: Gu, Y. ef al., (1997) Science: 206-209) There is eí al. (1992) Hum 3: Harlow and Lane, Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Press, 1990. Hawkins et al. (1992) 226: 889-896 Hezareh, M., et. al., (2001) J. Virology, 75 (24): 12161-12168 Holliger, P., ef al. (1993) Proc. Nati Acad. Sci. USA 90: Hoogenboom et al. (1991) 19: Hoogenboom H. R., (1997) Tech. 15: 62-70 and Chames P. (2000) Immunology Today 21: 371-378 Huston et al. (1988) Proc. Nati Acad .: 5879-5883 Hoshino K., et al (1999) J. Immunol. 162: 5041-5044 et al. (1989) Science 246: 1275-1281 Johnnson, B., (1991) Anal. 198: Johnsson, B., ef al. (1995) J. Mol .: 125-131 Jonsson, U., et al. (1991) Biotechniques 11: 620-627 Kanakaraj P., (1999) J. Exp. Med. 189: Kaufman, R. J. and Sharp, P. A., (1982) Mol. Biol. 159: 601-621 Kearney et al, J. Immunol. 123,1979, 1548-1550 S. A. and Green L. L. (2002) Current Opinion in Biotechnology 13: 593-597 K., et al (1999) Eur. J. Immunol.29: 2819-2825 Konishi, K., et al (1997) 209: 187-191 Kipriyanov, S.M., (1994) Mol. Immunol.31: 1047-1058 Kipriyanov, S.M., ef al. (1995) Human Antibodies and Hybridomas 6: 93-101 Leung, B. P., et al. (2001) J. Immunol. 167: 2879-2886 Liííle M. eí al 21: 364-370 BioTechniques Press. Wesíborough, MA. pp. (ISBN 1- 881299-21-X). Lund, J. ef al., J. Immunology (1991) 147: 2657-2662 McCafferiy et al.
Nafure (1990) 348: 552-554 Mclnnes, I. B. et. to the. (2000) Immunology Today 21: 312-315; Méndez et al., Nature Genetics.15: 146-156 (1997) Hoogenboom et al. (1991) 19: Hoogenboom H. R., (1997) Tech. 15: 62-70 and Chames P. (2000) Immunology Today 21: 371-378 Huston et al. (1988) Proc. Nati Acad .: 5879-5883 Hoshino K .; et al (1999) J. Immunol. 162: 5041-5044 et al. (1989) Science 246: 1275-1281 Johnnson, B., (1991) Anal. 198: Johnsson, B., ef al. (1995) J. Mol .: 125-131 Jonsson, U., ef al. (1991) Biotechniques 11: 620-627 Kanakaraj P., (1999) J. Exp. Med. 189: Kaufman, R. J. and Sharp, P. A., (1982) Mol. Biol. 159: 601-621 Kearney et al, J. Immunol. 123,1979, 1548-1550 S. A. and Green L. L. (2002) Current Opinion in Biotechnology 13: 593-597 K., et al (1999) Eur. J. Immunol.29: 2819-2825 Konishi, K., et al (1997) 209: 187-191 Kipriyanov, S.M., (1994) Mol. Immunol.31: 1047-1058 Kipriyanov, S.M., ef al. (1995) Human Antibodies and Hybridomas 6: 93-101 Leung, B. P., et al (2001) J. Immunol. 167: 2879-2886 Litíle M. eí al 21: 364-370 BioTechniques Press. Wesfborough, MA. pp. (ISBN 1 - 881299-21 -X). Lund, J. ef al. , J. Immunology (1991) 147: 2657-2662 McCaffery ef Naíure (1990) 348: 552-554 Mclnnes, I. B. eí. to the. (2000) Immunology Today 21: 312-315; Méndez ef al. , Naíure Geneíics 15: 146-156 (1997) Mizushima, S. and Nagata, S., (1990) Nucleic acids Research Vol 18, No. 17 Nakanishi, K. et al (2001) Ann. Rev. 1 9: Nakanishi K., et al (2001) Cytokine and Growth Factor Rev. 12: 53-72 Neeta, M. G., et al (2000) J. Immunol. 164: 2644-2649 Ober, R. J., et al (2001) Int. Immunol. 13: 1551-1559 Poljak, R. J., ef al. (1 994) Struclure: 1 121 -1 123 Seidman, J. G., Smiíh, J. A., and Síruhl eds; Wiley Interscience, N. Y., N. Y. (1 990) Sims, J. E., (2002) 14: 1 17-122 Sugawara, S. et al., (2001) 167.6568-6575 Sustained and Conlrolled Reléase Drug Delivery Sysiems, J. R. Robinson, Marcel Dekker, Inc., New York, 1978. Takeda, K., et al. (1 998) Immunily 8: 383-390 Taylor, L. D., et al. (1992) Nucí. Acids Res. 20: 6287-6295 Tissi L., eí al (1999) Infecí. Immunol. 67: 4545-50 T reníham, D. E. eí al (1977) J. Exp. Med. 146: Tsutsui, H. et al., (1999) Immunity 1 1: 359-67 Uriaub and Chasin, (1980) Proc. Nati Acad. Sci. USA 77: 4216-4220 S., ef al. (1996) J. Immunol. 156: 4274-4279 Ward et Naure 341: 544-546 Wei, X. Q., al (2000) J. Immunol. 166: 517-521 Winnacker, E. L. From Genes To Clones: Introduction To Gene Technology (1987) VCH Publishers, NY (translated by Horst 634 pp.
(ISBN Lund, J. ef al., J. Immunology (1991) 147: 2657-2662 McCafferty et al.
Nature (1990) 348: 552-554 Mclnnes, I. B. et. to the. (2000) Immunology Today 21: 312-31 5; Méndez ef al. , Nature Genetics 15: 146-156 (1997) Mizushima, S. and Nagata, S., (1990) Nucleic acids Research Vol 18, No. 17 Nakanishi, K. et al (2001) Ann. Rev. 19: Lund, J. ef al. , J. Immunology (1991) 147: 2657-2662 McCafferty et Nature (1990) 348: 552-554 Mclnnes, I. B. et. to the. (2000) Immunology Today 21: 312-315; Méndez ef al. , Nature Genetics 15: 146-156 (1997) Mizushima, S. and Nagata, S., (1990) Nucleic acids Research Vol 18, No. 17 Nakanishi, K. et al (2001) Ann. Rev. 19: Nakanishi K., et al (2001) Cytokine and Growth Factor Rev. 12: 53-72 Neeta, M. G., et al (2000) J. Immunol. 164: 2644-2649 Ober, R. J .. ei al (2001) Iní. Immunol. 13: 1 551-1559 Poljak, R. J., et al. (1994) Strucíure: 1 121 -1 123 Seidman, J. G., Smiíh, J. A., and Síruhl eds; Wiley Iníerscience, N. Y., N. Y. (1990) Sims, J. E., (2002) 14: 1 17-122 Sugawara, S. et al., (2001) 167.6568-6575 Sustained and Controlled Reverse Drug Delivery Systems, J. R. Robinson, Marcel Dekker, Inc., New York, 1 978. Takeda, K., ef al. (1,998) I mmuniíy 8: 383-390 Taylor, L. D., ef al. (1 992) Nucí. Acids Res. 20: 6287-6295 Tissi L., at (1,999) Infecí. Immunol. 67: 4545-50 T reníham, D. E. eí al (1977) J. Exp. Med. 146: Nakanishi K., al (2001) Cyíokine and Growíh Faclor Rev. 12: 53-72 Neeta, M. G., et al (2000) J. I mmunol. 164: 2644-2649 Ober, R. J., et al (2001) Int. Immunol. 13: 1 551-1559 Poljak, R. J., et al. (1994) Strucíure: 1 121 -1 123 Seidman, J. G., Smiíh, J. A., and Síruhl eds; Wiley Iníerscience, N. Y., N. Y. (1990) Sims, J. E., (2002) 14: 1 17-122 Sugawara, S. eí al., (2001) 167,6568-6575 Susíained and Conírolled Reléase Drug Delivery Sysiems, J. R. Robinson, Marcel Dekker, Inc., New York, 1 978. Takeda, K., ef al. (1998) Immuniíy 8: 383-390 Taylor, L. D., ef al. (1 992) Nucí. Acids Res. 20: 6287-6295 Tissi L., eí al (1999) Infecí. Immunol. 67: 4545-50 T reníham, D. E. eí al (1977) J. Exp. Med. 146: In spite of the fact that a number of modalities and characteristics have been described earlier, it will be understood for those experts in the field that the modifications and variations of the described and characterizing modalities can be made without departing from the present description or invention as defined in the attached claims. Each of the publications mentioned herein is incorporated for reference.

Claims (79)

  1. CLAIMS 1. A binding protein comprising an antigen binding domain capable of binding human IL-18, said antigen-binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of: CDR-H1. (SEQ I D NO: 42), where; X2 is Y, G, R, S, or C; X3 is W, G, Y, D, S, V, or I; X4 is H, W, Y, M, L, or D; X5 is G, Y, S, N, or H; X6 is W, or does not occur; and X7 is T, S, G, or does not occur; CDR-H2. X? -X2-X3-X -X5-X6-X -X8-Xg-X? O-? ? -X? 2-Xi3-X? -Xi5-X .6-X .7 (SEQ ID NO: 43), wherein; Xn is F, Y, H, S, or V; X2 is or F; X3 is Y, S, or W; X4 is P, Y, or S; X5 is G, S, R, or D; X6 is D, or G; X7 is S, T, G, or R; X8 is E, T, I or N; X9 is T, Y, N, K, or H; X10 is R, Y, or S; Xn is Y, N, or S; X12 is S, P, A, or V; X13 is P, S, or D; X14 is T, L, or S; X15 is F, K, or V; X16 is Q, S, or K; and X17 is G, or does not occur; CDR-H3. X- | -X2-X3- -X5- 6-X7- 8-X9-Xl0"X 11" X .2"X .3 ~ l4" l 5"Xl í X17-X18 (SEQ ID NO: 44), wherein; X! is V, D, E, S, or C; X2 is G, R, D, S, K, L, Y, or A; X3 is G, Y, R; X4 is G, S, Y, N, T, or D; X5 is W, S, A, G, Y, or T; X6 is Y, G, S, F, W, or N; X7 is P, S, F, Y, V, G, W, or V; X8 is Y, F, D, P, M, I or N; X9 is T, W, D, L, Y, E, P, F, or G; X10 is F, D, Y, H, V, Y, or does not occur; X11 is D, Y, F, L, or does not occur; X12 is I, D, Y, or does not occur; X-i3 is Y, or does not occur; X? 4 is Y, or does not occur; X15 is G, or does not occur; X-I6 is M, or does not occur; X-? 7 is D, or does not occur; and X18 is V, or does not occur; CDR-L1. X -? - X2- 3-X-5-6-7-9-? Or "?? - -? 2-Xi3 ~ i4- i5-X.6-X 17 (SEQ: 45), where XI is R, or K, X2 is A, G, or S, X3 is S, X4 is E, R, Q, or H, X5 is S, I, T, or N, X6 is I, V, L , or F; X7 is S, G, L, N, or R; X8 is S, G, Y, R, N, H, or D; X9 is N, G, Y, R, or S; X10 is L , Y, S, or D, XII is A, L, N, V, G, or D, X12 is A, N, E, K, G, or does not occur, X-13 is K, T, N, or does not occur, X1 is N, Y, T, or does not occur, X15 is Y, L, or does not occur, X16 is L, C, Y, or does not occur, and X17 is A, D, or does not occur, CDR-L2, X ^ Xz-Xa-X ^ Xs-Xe-X? (SEQ NO: 46), where X is T, G, S, W, or E, X2 is A, V, T, I, or L; X3 is S, or F; X4 is T, I, N, S, R, or Y; X5 is R, or L; X6 is A, Q, E, or F; and X7 is T, or S; and CDR-L3. (SEQ ID NO: 47), wherein; X, is Q or M; X2 is Q, H, or Y; X3 is Y, N, G, S, or R; X4 is N, H, Y, D, G, V, L, or I; X5 is G, I, Y, S, Q, F, or E; X6 is W, S, T, L, I, or F; X7 is P, L, T, D, or I; X8 is S, L, P, C, W, I or F; X9 is I, T, S, or does not occur; and X10 is T, or does not occur. The binding protein according to claim 1, wherein said at least one CDR comprises an amino acid sequence selected from the group consisting of: Residues 31-35 of SEQ ID NO: 6; Residues 50-66 of SEQ ID NO .: 6; Residues 99-110 of SEQ ID NO .: 6; Residues 24-34 of SEQ ID NO .: 7; Residues 50-56 of SEQ ID NO .: 7; Residues 89-98 of SEQ ID NO .: 7; Residues 31-37 of SEQ ID NO .: 8; Residues 52-67 of SEQ ID NO .: 8; Residues 100-110 of SEQ ID NO .: 8; Residues 24-35 of SEQ ID NO .: 9; Residues 21-27 of SEQ ID NO .: 9; Residues 90-98 of SEQ ID NO .: 9; Residues 31-35 of SEQ ID NO .: 10; Residues 50-65 of SEQ ID NO .: 10; Residues 98-107 of SEQ ID NO .: 10; Residues 24-34 of SEQ ID NO .: 11; Residues 50-56 of SEQ ID NO .: 11; Residues 89-97 of: 11; Residues 31-37 of: 12; Residues 52-67 of SEQ ID NO .: 12; Residues 100-108 of SEQ ID NO .: 12; Residues 24-35 of SEQ ID NO .: 13; Residues 51-57 of SEQ ID NO: 13; Residues 90-98 of SEQ ID NO .: 13; Residues 31-35 of SEQ ID NO .: 14; Residues 50-66 of SEQ ID NO .: 14; Residues 99-111 of SEQ ID NO .: 14; Residues 24-40 of SEQ ID NO .: 15; Residues 56-62 of SEQ ID NO: 15; Residues 95-103 of SEQ ID NO .: 15; Residues 31-37 of SEQ ID NO .: 16; Residues 52-67 of SEQ ID NO .: 16; Residues 100-109 of SEQ ID NO .: 16; Residues 24-35 of SEQ ID NO .: 17; Residues 51-57 of SEQ ID NO .: 17; Residues 90-98 of SEQ ID NO .: 17; Residues 31-35 of SEQ ID NO .: 18; Residues 20-36 of SEQ ID NO .: 18; Residues 99-108 of SEQ ID NO .: 18; Residues 24-34 of SEQ ID NO .: 19; Residues 50-56 of SEQ ID NO .: 19; Residues 89-97 of SEQ ID NO .: 19; Residues 31-35 of SEQ ID NO: 20; Residues 52-67 of SEQ ID NO: 20; Residues 100-108 of SEQ ID NO .: 20; Residues 24-35 of SEQ ID NO .: 21; Residues 51-57 of SEQ ID NO .: 21; Residues 90-98 of SEQ ID NO .: 21; Residues 31-35 of SEQ ID NO .: 22; Residues 50-66 of SEQ ID NO .: 22; Residues 99-116 of SEQ ID NO .: 22; Residues 24-39 of SEQ ID NO .: 23; Residues 55-61 of SEQ ID NO: 23; Residues 94-102 of SEQ ID NO .: 23; Residues 31-37 of SEQ ID NO .: 24; Residues 52-67 of SEQ ID NO .: 24; Residues 100-109 of SEQ ID NO .: 24; Residues 24-35 of SEQ ID NO .: 25; Residues 51-57 of SEQ ID NO .: 25; Residues 90-98 of SEQ ID NO .: 25; Residues 31-37 of SEQ ID NO .: 26; Residues 52-67 of SEQ ID NO .: 26; Residues 100-109 of SEQ ID NO .: 26; Residues 24-35 of SEQ ID NO .: 27; Residues 51-57 of SEQ ID NO: 27; Residues 90-98 of SEQ ID NO .: 27; Residues 31-37 of SEQ ID NO .: 28; Residues 52-67 of SEQ ID NO .: 28; Residues 100-108 of SEQ ID NO .: 28; Residues 24-35 of SEQ ID NO .: 29; Residues 51-57 of SEQ ID NO .: 29; Residues 90-98 of SEQ ID NO .: 29; Residues 31-37 of SEQ ID NO .: 30; Residues 52-67 of SEQ ID NO: 30; Residues 99-109 of SEQ ID NO: 30; Residues 24-35 of SEQ ID NO .: 31; Residues 51-57 of SEQ ID NO: 31; Residues 90-98 of SEQ ID NO .: 31; Residues 31-37 of SEQ ID NO .: 32; Residues 52-67 of SEQ ID NO .: 32; Residues 100-109 of SEQ ID NO .: 32; Residues 24-35 of SEQ ID NO .: 33; Residues 51-57 of SEQ ID NO .: 33; Residues 90-98 of SEQ ID NO .: 33; Residues 31-37 of SEQ ID NO .: 34; Residues 52-67 of SEQ ID NO .: 34; Residues 100-108 of SEQ ID NO .: 34; Residues 24-35 of SEQ ID NO .: 35; Residues 51-57 of SEQ ID NO: 35; Residues 90-98 of SEQ ID NO .: 35; Residues 31-35 of SEQ ID NO .: 36; Residues 50-66 of SEQ ID NO .: 36; Residues 99-116 of SEQ ID NO .: 36; Residues 24-39 of SEQ NO .: 37; Residues 55-61 of SEQ ID NO .: 37; Residues 94-102 of SEQ ID NO .: 37; Residues 31-35 of SEQ ID NO .: 38; Residues 50-66 of SEQ ID NO .: 38; Residues 99-108 of SEQ NO .: 38; Residues 24-35 of SEQ ID NO .: 39; Residues 51-57 of SEQ ID NO .: 39; Residues 90-98 of SEQ ID NO .: 39; Residues 31-37 of SEQ ID NO .: 40; Residues 52-67 of SEQ ID NO .: 40; Residues 97-109 of SEQ ID NO .: 40; Residues 24-40 of SEQ ID NO .: 41; Residues 56-62 of SEQ ID NO .: 41; and Residues 95-103 of SEQ ID NO: 41. 3. The binding protein according to claim 2 wherein said binding protein comprises at least 3 CDRs. 4. The binding protein according to claim 2 wherein said antigen-binding domain comprises a VH. 5. The binding protein according to claim 4 wherein said VH comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 6; SEQ ID NO: 8; SEQ ID NO: 10; SEQ ID NO: 12; SEQ ID NO: 14; SEQ ID NO: 16; SEQ ID NO: 18; SEQ ID NO: 20; SEQ ID NO: 22; SEQ ID NO: 24; SEQ ID NO: 26; SEQ ID NO: 28; SEQ ID NO: 30; SEQ ID NO: 32; SEQ ID NO: 34; SEQ ID NO: 36; SEQ ID NO: 38; and SEQ ID NO: 40. The binding protein of claim 2, wherein said antigen-binding domain comprises a VL. The binding protein according to claim 6 wherein said V comprises an amino acid sequence selected from the group consisting of SEQ I D NO: 7; SEQ I D NO: 9; SEQ NO: 1 1; SEQ I D NO: 13; SEQ ID NO: 15; SEQ ID NO: 17; SEQ I D NO: 1 9; SEQ I D NO: 21; SEQ I D NO: 23; NO: 25; SEQ I D NO: 27; SEQ I D NO: 29; SEQ I D NO: 31; SEQ I D NO: 33; SEQ ID NO: 35; SEQ I D NO: 37; SEQ ID NO: 39; and SEQ I D NO: 41. 8. The binding protein according to claim 2 wherein said antigen-binding domain comprises a VH and a VL. The binding protein according to claim 7 further comprising a VH wherein said VH comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 6; SEQ ID NO: 8; SEQ ID NO: 10; SEQ ID NO: 12; SEQ ID NO: 14; SEQ ID NO: 16; SEQ ID NO: 18; SEQ ID NO: 20; SEQ ID NO: 22; SEQ ID NO: 24; SEQ ID NO: 26; SEQ ID NO: 28; SEQ ID NO: 30; SEQ ID NO: 32; SEQ ID NO: 34; SEQ ID NO: 36; SEQ I D NO: 38; and SEQ ID NO: 40. 1 0. The binding protein according to claim 8 wherein said VL comprises an amino acid sequence of SEQ ID NO. : 7 and said VH comprises an amino acid sequence of SEQ I D NO. : 6. 1 1. The binding protein according to claim 2, further comprising a domain consisting of a heavy chain immunoglobulin selected from the group consisting of a human IgM-like domain; a domain consists of human IgG 1; a domain consists of human IgG2; a human LGG3 consanct domain; a domain constanie IgG4 human; a domain consisting of human IgE and a human IgA constant domain. The binding protein according to claim 1, wherein said heavy chain immunoglobulin constant region domain is a human IgG 1 consigning domain. The binding protein according to claim 12, wherein said domain comprises human IgG1 comprises amino acid sequence selected from the group consisting of: SEQ ID NO: 2, and SEQ ID NO. :3. The binding protein according to claim 2, further comprising a domain consisting of light chain immunoglobulin selected from the group consisting of: a human Ig kappa constant domain; and a domain consisting of human Ig lambda. 15. The binding protein according to claim 14, wherein said light chain immunoglobulin constant region domain is a human kappa lg constant domain comprising amino acid sequence SEQ ID NO: 4. The binding protein according to claim 14, wherein said region domain consists of light chain immunoglobulin is a human Ig lambda constant domain comprising amino acid sequence SEQ ID NO: 5. 17. The binding protein according to claim 2, wherein said binding protein is selected from the group consisting of: an immunoglobulin molecule; a scFv; a monoclonal antibody; a human antibody; a chimeric antibody; a humanized antibody; a single domain antibody; a Fab fragment; a Fab 'fragment; an F (ab ') 2; an Fv; and a disulfide-linked Fv. The binding protein according to claim 17, wherein said binding protein is a human antibody. 19. A binding protein capable of binding human IL-18, said binding protein comprising: a heavy region consisting of Ig having an amino acid sequence selected from the group consisting of: SEQ ID NO: 2 and SEQ ID NO: 3; a light region consisted of Ig having an amino acid sequence of the group consisting of: SEQ ID NO: 4 and SEQ ID NO: 5; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 6,, and a light variable region Ig having an amino acid sequence of SEQ ID NO: 7. 20. A binding protein capable of binding human L-1 8, said binding proiein comprising: a heavy Ig-containing region having an amino acid sequence of SEQ ID NO: 3; a constant light region IG having an amino acid sequence of SEQ ID NO: 4; a variable heavy region Ig having an amino acid sequence of SEQ ID NO: 6; and a light variable region Ig having an amino acid sequence of SEQ ID NO: 7. twenty-one . A neutralizing binding protein, wherein said neutralizing binding protein comprises a binding protein according to any one of claims 1-20, and wherein said binding protein is capable of neutralizing I L-1 8. Neutralizing binding protein according to claim 12, wherein said IL-18 is selected from the group consisting of pro-human IL-1; I L-18 human mature and I L-1 8 human truncated. 23. The neutralizing binding protein according to claim 21, wherein said neutralizing binding protein decreases the ability of L-1 8. to be added to its receptor. 24. The neutralizing binding protein according to claim 23, wherein said neutralizing linking protein decreases the ability of pro-human IL-1 8, mature human I-1-8, or bound human 1-alpha-18 to bind to its receptor. . 25. The neutralizing binding protein according to claim 21, wherein said neutralizing binding protein is capable of reducing one or more biological activities I L-1 8 selected from the group consisting of: Th 1 modulation; Th2 modulation; Nk modulation; neutrophil modulation; monocyte-macrophage lineage modulation; pneumophilic modulation; eosinophil modulation; B cell modulation; modulation of cyclokine; chemokine modulation; adhesion molecule modulation; and modulation of recruitment of cells. 26. The neutralizing binding protein according to claim 21, wherein said neutralizing binding protein has a dissociation constancy (KD) selected from the group consisting of: more than about 10"7 M, to more than about 1 0" 8 M; to more than approximately 10"9 M, to more than approximately 1 0" 10 M; more than approximately 10"11 M; more than approximately 1 0" 12 M; and more than about 10"13M 27. The neutralizing binding protein according to claim 21, wherein said neutralizing binding protein has an active scale selected from the group consisting of: at least about 102M" 1s "1, at least about 103M "V; at least about 104M "V1, at least about 1MM" 1S "1, and at least about 106M" 1S "1. 28. The neufralizing binding protein according to claim 21, wherein said binding protein neutralizes an inactive scale. selected from the group consisting of: more than approximately 10"3s" 1, more than approximately 10"4s" 1, more than approximately 1 0"5s" 1, and more than approximately 1 0" -1 29. A labeled binding protein comprising a binding protein of any one of claims 1-20, wherein said binding protein is conjugated to a detectable label. 30. The labeled binding proiein according to claim 29, wherein the detectable label is selected from the group consisting of a radiochemistry, an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and bioin. 31 The labeled binding prolein according to claim 30, wherein said tag is a radioetiquey selected from the group consisting of: 3H, 14C, 35S, 90Y, "Te, 1 1 1 In, 125 I, 131 L, 177 Lu, 166 Ho, or 53. A conjugated binding protein comprising a binding protein of any one of claims 1-20, wherein said binding protein is conjugated to a cyclooxy or diaepatric agent. , wherein said therapeutic or cytotoxic agent is selected from the group consisting of: an anti-metabolism, an alkylation agent, an antibiotic, a growth factor, a cyclokine, an ani-angiogenic agent, an antimicotic agent, an anthracycline, toxin, and an apoptotic agent 34. An asylated nucleic acid encoding a binding protein amino acid sequence of any one of claims 1 - 20. 35. A vector comprising an acid isolated nucleic acid according to claim 34. 36. The neighbor according to claim 35 wherein said neighbor is selected from the group consisting of: pcDNA, pTT, pTT3, pEFBOS, pBV, pJV, and pBJ. 37. A host cell comprising a neighbor according to any of claims 35 or 36. 38. The host cell according to claim 37, wherein said host cell is a prokaryotic cell. 39. The host cell according to claim 38, wherein said host cell is E. coli. 40. The host cell according to claim 37, wherein said host cell is a eukaryotic cell. 41 The host cell according to claim 40, wherein said eukaryotic cell is selected from the group consisting of prolific cell, animal cell, pineapple cell and fungal cell. 42. The host cell of claim 41, wherein said eukaryotic cell is an animal cell selected from the group consisting of: a mammalian cell, a bird cell, and an insect cell. 43. The host cell according to claim 42, wherein said mammalian cell is a CHO cell. 44. The host cell according to claim 42, wherein said host cell is COS. 45. The host cell according to claim 41, wherein said eukaryotic cell is Saccharomyces cerevisiae. 46. The host cell according to claim 42, wherein said animal cell is an insemination Sf9 cell. 47. A method for producing a binding proiein binding human IL-1, comprising culturing the host cell of any of claims 37-46 in a culture medium under conditions sufficient to produce a binding protein that binds I L -1 8 human. 48. A binding protein produced according to the method of claim 47. 49. A crystallized binding protein comprising a binding protein according to any of claims 1 -28, wherein said binding protein exists as a crystal. 50. The crystallized binding protein according to claim 49, wherein said crystal is a vehicle-free pharmaceutical controlled release crystal. 51 The crystallized binding protein according to claim 49, wherein said binding protein has a longer half-life in vivo than the soluble protein of said binding protein. 52. The chiralized binding protein according to claim 49, wherein said binding prolein retains the biological activity. 53. A composition for the release of a binding protein of said composition comprising: (a) a formulation, wherein said formulation comprises a chiralized binding protein, according to any of claims 49-52, and an active ingredient.; and (b) at least one polymeric vehicle. 54. The composition according to claim 53, wherein said polymer carrier is a polymer selected from one or more of the group consisting of: poly (acrylic acid), poly (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-go-glycolic acid) or PLGA, poly (b-hydroxybutyrate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organo) phosphazene); poly (orioles), poly (vinyl alcohol), poly (vinylpyrrolidone), copolymers of vinyl alkyl-maleic anhydride, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelaine, hyaluronic acid, oligosaccharides, glycolino glycans, sulfated polysaccharides, mixtures and copolymers thereof. 55. The composition according to claim 53, wherein said ingredient is selected from the group consisting of albumin, sucrose, irthalosa, lactitol, gelatin, hidoxypropyl-β-cyclodextrin, methoxypolyethylene glycol and polyethylene glycol. 56. A method for bringing a mammal comprising the step of administering to the mammal an effective canine of the composition according to claim 53. 57. A method for regulating gene expression of a gene of interest comprising the steps of (a) providing an IL-18 modulator, and (b) contacting said modulate to a cell wherein said gene of interest is selected from the group consisting of Genbank identification numbers: 58. The method according to claim 57, wherein said modulator is an antagonist. 59. The method according to claim 57 wherein said modulator is IL-1 8. 60. The method according to claim 57 wherein said modulator is selected from the group consisting of a binding protein according to any of claims 1 -28. 61 A pharmaceutical composition comprising the binding protein according to any of claims 1 - 28, and a pharmaceutically acceptable carrier. 62. The pharmaceutical composition according to claim 61 further comprising at least one additional therapeutic agent for bringing a disorder in which the activity of IL-18 is deleterious. 63. The pharmaceutical composition according to claim 62, wherein said additional agent is selected from the group consisting of: angiogenesis inhibitors; kinase inhibitors; co-stimulation molecule blockers; adhesion molecule blockers; anti-cytokine antibody or functional fragment thereof; metoirexaío; chorioicosteroids; cyclosporin; rapamycin; FK506; and non-inflammatory and non-spheroidal agents. 64. A method for reducing the activity of human L-1 8 comprising human IL-18 with the binding protein according to any one of claims 1 to 28 of the invention that the activity of human IL-1 8 is reduced. 65. A method for reducing the activity of human I L-18 in a human subject suffering from a disorder in which the activity of I L-1 8 is harmful, comprising administering to the human subject the binding protein according to any of claims 1 -28 in such a way that the activity of human I L-1 8 in the human subject is reduced. 66. A method for treating a subject for a disease or disorder in which the activity of IL-18 is deleterious when administering to the subject the binding protein according to any of claims 1 -28 in such a way that the treatment is achieved. The method according to claim 66, wherein said disorder is selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile chronic aririitis, Lyme's aryrisis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropary, sysmal lupus erythematosus, Chron's disease , ulcerative colitis, inflammatory inflammatory disease, insulin-dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, scleroderma dermatitis, graft against host disease, rejection of organ transplany (including but not limited to bone marrow and solid organ rejection) ), chronic or acute immune disease, disease associated with organ transplantation, sarcoidosis, arteriosclerosis, disseminated vascular coagulation, Kawashi disease, Grave disease, nephrotic syndrome, chronic fatigue syndrome, Wegener granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic acivive hepatitis, uveiitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, chorea Hunfingion, Parkinson's disease, Alzheimer's disease, primary biliary cirrhosis , hemolytic anemia, malignancy, heart failure, myocardial infarction, Addison's disease, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt's syndrome, respiratory distress syndrome (acute), alopecia, alopecia areala, ulcerative eolithic aryropathy, synovitis enyerophalic, chlamydia, yersinia and salmonella associated with aryropaphy, spondylar papilloma, aferomaiosa / aryeriosclerosis disease, atopic allergy, auino-immune bolus disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, auinoimmune hemolytic anemia, Coombs positive hemolytic anemia, acquired pernicious anemia, juvenile pernicious anemia, myalgic encephalitis / Royal Free disease, chronic mucocutaneous candidiasis, giganie cell arteritis, primary sclerosis hepaiitis , Cryptogenic autoimmune hepaitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common variable immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease , alveoliíos of cryptogenic fibrosis, post-inflammatory infersíicial lung disease, iníersíicial pneumonitis, iníersíicial lung disease associated with connective liver disease, lung disease associated with kidney disease mixed ectivity, infersiicial lung disease associated with sysiomemic sclerosis, inflammatory lung disease associated with rheumatoid arthritis, lung disease associated with systemic lupus erythematosus, lung disease associated with dermamyosilitis / polymyositis, lung disease associated with Sjögren's disease, lung disease associated with ankylosing spondylitis, diffuse vasculiic lung disease, lung disease associated with hemosiderosis, drug-induced iniersficial lung disease, radiation fibrosis, bronchiolitis oblières, chronic eosinophilic pneumonia, lymphocytic infillraine lung disease, post-infectious interstitial lung disease , Aririíis de goía, auinoimmune hepatitis, auioimmune hepatitis, hepatitis auioimmune type 1 (lupoid or classic hemopathy hepaitis), autoimmune hepatitis type 2 (anti-LKM antibody hepatitis), immune-mediated hypoglycemia, resistance Type B insulin with acaniosis nigricans, hypoparathyroidism, acute autoimmune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosis cholangitis, type 1 psoriasis, type 2 psoriasis, idiomatic leucopaenia, autoimmune neutropenia , NOS renal disease, NOS, glomerulonephritis, microscopic vasculitis of the kidneys, Lyme disease, discoid erythematosus lupus, male idiomatic infertility or NOS, sperm immunoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hyper-tension secondary to tissue disease connective, Goodpasiure syndrome, pulmonary manifestation of polyacryllitis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjögren's syndrome, Takayusu's disease / arteritis, autoimmune ombrombocytopenia, idiomatic írombocytopenia, ileoid disease nymphoid, hyperhyroidism, auimmune hypothyroidism (Alzheimer's disease), aurophically auioimmune hypoiyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo, acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, coleostasis, disease of idiosyncratic liver, drug-induced hepatitis, non-alcoholic disease, allergy and asthma, group B (GBS) infection, menia disorders (eg, depression and schizophrenia), Type Th1 and Type Th2-mediated diseases, acute and chronic pain, and cancer. 68. A method to eradicate a patient who suffers from a disorder in which IL-18 is harmful by understanding the stage of administering the protein. binding according to any of claims 1 to 28 before, concurrent, or after administration of a second agent, wherein the second agent is selected from the group consisting of an antibody, or fragment thereof, capable of binding human IL-12; methotrexaph; an antibody, or fragment thereof, capable of binding human TNF; chorioicosteroids, cyclosporine, rapamycin, FK506, and nonsteroidal anti-inflammatory agents. 69. A neuiralizing binding protein wherein said neutralizing linker is able to bind mature human IL-18, but not specifically bind pro-human IL-1, and wherein said neutralizing binding protein is selected from the group consisting of a human antibody; a chimeric animal; a humanized antibody and a CDR-grafted antibody. 70. The binding protein according to claim 1, wherein said binding protein is capable of binding mature human IL-18, but not specifically binding pro-human IL-18. 71 A neutralizing binding protein wherein said neutralizing binding protein is capable of competing with 125-2H antibody to bind human IL-18, and wherein said neutralizing binding protein is selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and a CDR-grafted antibody. 72. The binding protein according to claim 1, wherein said binding protein is capable of competing with the 125-2H antibody to bind human L-1 8. 73. A neutralizing binding protein wherein said neutralizing binding protein is not capable of competing with the 125-2H antibody to bind human IL-18, and wherein said neutralizing binding protein is selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and a CDR-grafted antibody. 74. The binding protein according to claim 1, wherein said binding protein is not capable of competing with the 125-2H antibody to bind human L-18. 75. A neutralizing binding protein wherein said neutralizing protein is not capable of competing with a binding protein selected from the group consisting of antibody 2.5 (E) mg 1, and IL-18BP to bind human IL-18, and wherein said Neutralizing binding prolein is selected from the group consisting of a human antibody; a chimeric antibody; a humanized antibody and an antibody injected with CDR. 76. The binding protein according to claim 1, wherein said binding protein is not capable of competing with a binding protein selected from the group consisting of antibody 2.5 (E) mg 1, and IL-1 8BP to bind human 1L-18 . 77. The binding protein according to claim 8 wherein said V comprises an amino acid sequence of SEQ ID NO: 9, and said VH comprises an amino acid sequence of SEQ ID NO: 8. 78. A binding protein capable of binding human L-18, said binding protein comprising: an Ig-containing heavy region having an amino acid sequence selected from the group consisting of SEQ ID NO: 2 and SEQ ID NO: 3; a light region consiiates IG having an amino acid sequence of the group consisting of SEQ ID NO: 4 and SEQ I D NO: 5; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 8, and a light variable region Ig having an amino acid sequence of SEQ ID NO: 9. 79. A binding protein capable of binding human IL-18, said binding protein comprising: a heavy region comprising Ig having an amino acid sequence selected from the group consisting of SEQ ID NO: 3; a constant light region IG having an amino acid sequence of the group consisting of SEQ ID NO: 4; a variable heavy region Ig having an amino acid sequence of SEQ I D NO: 8, and a light variable region Ig having an amino acid sequence of SEQ ID NO: 9.
MXPA/A/2006/005469A 2003-11-12 2006-05-12 Il-18 binding proteins MXPA06005469A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10706689 2003-11-12

Publications (1)

Publication Number Publication Date
MXPA06005469A true MXPA06005469A (en) 2006-10-17

Family

ID=

Similar Documents

Publication Publication Date Title
JP5054977B2 (en) IL-18 binding protein
US7968684B2 (en) IL-18 binding proteins
US8629257B2 (en) IL-12/p40 binding proteins
US8383778B2 (en) IL-1 binding proteins
US20120275996A1 (en) IL-1 Binding Proteins
US20110165063A1 (en) Il-1 binding proteins
AU2011224023B2 (en) IL-18 binding proteins
MXPA06005469A (en) Il-18 binding proteins