AU2011224023B2 - IL-18 binding proteins - Google Patents

IL-18 binding proteins Download PDF

Info

Publication number
AU2011224023B2
AU2011224023B2 AU2011224023A AU2011224023A AU2011224023B2 AU 2011224023 B2 AU2011224023 B2 AU 2011224023B2 AU 2011224023 A AU2011224023 A AU 2011224023A AU 2011224023 A AU2011224023 A AU 2011224023A AU 2011224023 B2 AU2011224023 B2 AU 2011224023B2
Authority
AU
Australia
Prior art keywords
antibody
seq
antigen binding
binding portion
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2011224023A
Other versions
AU2011224023A1 (en
AU2011224023C1 (en
Inventor
John Badcook
Tariq Ghayur
Larry Green
Brad Hedberg
Xiao-Chi Jia
Jaspal Singh Kang
Boris Labkovsky
Jeffery W. Voss
James Wieler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
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
Priority claimed from AU2004290073A external-priority patent/AU2004290073B2/en
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to AU2011224023A priority Critical patent/AU2011224023C1/en
Publication of AU2011224023A1 publication Critical patent/AU2011224023A1/en
Publication of AU2011224023B2 publication Critical patent/AU2011224023B2/en
Priority to AU2012261666A priority patent/AU2012261666A1/en
Assigned to ABBVIE INC. reassignment ABBVIE INC. Request for Assignment Assignors: ABBOTT LABORATORIES
Application granted granted Critical
Publication of AU2011224023C1 publication Critical patent/AU2011224023C1/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

IL -18 BINDING PRO TEINS Abstract The present invention encompasses IL-1 8 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 hlL-18 and/or that neutralize hIL-1 8 activity in vitro and in vivo. An antibody of the io 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 hlL- 18 and for inhibiting hIL-18 activity, e.g., in a human subject suffering from a disorder in which hlL 18 activity is detrimental.

Description

S&F Ref: 762439D1 AUSTRALIA PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT Name and Address Abbott Laboratories, of D-377 AP6A/1, 100 Abbott Park of Applicant: Road, Abbott Park, Illinois, 60064, United States of America Actual Inventor(s): Boris Labkovsky Brad Hedberg James Wieler Jaspal Singh Kang Xiao-Chi Jia John Badcook Larry Green Tariq Ghayur Jeffery W. Voss Address for Service: Spruson & Ferguson St Martins Tower Level 35 31 Market Street Sydney NSW 2000 (CCN 3710000177) Invention Title: IL-18 binding proteins The following statement is a full description of this invention, including the best method of performing it known to me/us: 5845c(5588053_1) IL-18 BINDING PROTEINS Cross-reference to Related Application This application claims the benefit of priority to US application no. 10/706,689, filed S5 November 12, 2003. Field of the Invention The present invention relates interleukin 18 (IL-18) binding proteins, and specifically to their uses in the prevention and/or treatment of acute and chronic inflammatory diseases. Background of the Invention Interleukin-18 (IL-18) was originally described in 1989 as interferon-gamma inducing factor (IGIF) and is a pro-inflammatory cytokine with various functions in addition to an ability to induce interferon gamma. These biological properties include activation of NF-Kb, Fas ligand expression, the induction of both CC and CXC chemokines, and increased production of competent human immunodeficiency virus. Due to the ability of IL-18 to induce interferon gamma production in T cells and macrophages, it plays an important role in Th -type immune responses and participates in both innate and acquired immunity. IL-18 is related to the IL-I family in terms of both structure and function. For reviews of IL-18 structure, function and 0 biological activity, see for example Dinarello, C. et al. (1998) J. Leukoc. Biol. 63:658-654; Dinarello, C.A. (1999) Methods 19:121-132; and Dinarello, C.A. (1999) J. Allergy Clin. Immunol. 103:11-24; (McInnes, 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 18 kDa active form in endotoxin-stimulated cells by caspase 1 (Ghayur, T. et al., (1997) Nature 386:619-623; Gu, Y. et al., (1997) Science 275:206-209) and in Fas-L or bacterial DNA stimulated cells by caspases 4, 5 and 6 (Tsutsui, H. et al., (1999) Immunity 11:359-67; Ghayur, T., Unpublished Observations). Pro-IL-1 8 is also proteolytically processed by other proteases such as neutrophil proteinase 3 50 (Sugawara, S. et al., (2001) J. hnmunol., 167, 6568-6575 ), caspase 3 (Akita, K. et al., (1997) J. Biol. Chem. 272, 26595-26603 ), and serine proteases elastase and cathepsin (Gracie J. A., et al., (2003) Journal of Leukocyte Biology 73, 213-224). Both human and murine IL-18 lack a classical leader sequence and the mechanism of mature IL-18 release by cells is not well understood.
I
2 The biological activities of IL-18 are mediated through IL- 18 binding to a heterodimeric IL-I 8 receptor (IL-I 8R) that consists of two subunits: the a-subunit (a member of the IL-iR family, also termed IL-I R-related protein-I or IL-lRrpl) and the p-subunit (also termed IL-18R accessory proteins, IL-I 8AP or AcPL). The IL-1 8Ra subunit binds IL-1 8 directly, but is incapable of signal transduction. The 3-subunit does not bind IL-18 by itself, but in conjunction with the a-subunit forms the high affinity receptor (Kp = 0.3 nM) that is required for signal transduction (Sims, J.E., (2002) Current Opin Immunol. 14:117-122). IL-18 signal transduction via the IL-18Rap complex is similar to the IL-iR and Toll like receptor (TLR)'systems. IL-18R signalling uses the signal transduction molecules, such as MyD88, IRAK, TRAF6 and results in similar responses (e.g. activation of NIK, IkB kinases, NF-kB, JNK and p38 MAP kinase) as does IL-I. Requirement for IL-18Ra and signal transduction molecules in mediating IL-18 bioactivity has been confirmed using IL-I 8Rax subunit (Hoshino K., et al (1999) J. Immunol. 162:5041 5044;) MyD88 (Adachi O., et al. (1998) Immunity 9:143-150) or IRAK (Kanakaraj P., (1999) J. Exp. Med. 189:1129-1138) knockouts respectively. Antibodies that bind IL-18 are known in the art. Mouse antibodies capable of neutralizing IL-18 are disclosed in EP 0 974 600. Human antibodies to IL-1 8 have been disclosed in PCT publication WO 0158956 and incorporated herein by reference. The present invention provides a novel family of binding proteins, human antibodies, and fragments thereof, capable of binding IL-18, binding with high affinity, and binding and neutralizing IL-18. Summary of the Invention This invention pertains to IL- 18 binding proteins, particularly antibodies to human IL-18, as well as methods of making and using such binding proteins. One aspect of the invention pertains to a method of regulating gene expression using a modulator of IL-18. One aspect of this invention pertains to an isolated monoclonal antibody, or antigen binding portion thereof, capable of binding human IL-18, wherein the antibody, or antigen binding portion thereof, comprises: a heavy chain variable region comprising CDR], CDR2, and CDR3 sequences, and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, wherein the heavy chain variable region CDR3 sequence comprises an amino acid sequence of residues 99 110 of SEQ ID NO:6, or an amino acid sequence of residues 99-110 of SEQ ID NO:6 containing conservative amino acid substitutions.
2a 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. XI-X 2
-X
3
-X
4 --Xr-X6-X. (SEQ ID NO: 42), wherein; X, is S, N, H, R, or Y; X2 is Y, G, R, S. or C:
X
3 is W. G, Y, D, S, V, or 1;
X
4 is I, H, W, Y, M, L, or D; X5 is G, Y, S, N, or H;
X
6 is W, or is not present; and
X
7 is T, S, G, or is not present; CDR-H2. XI-X 2
-X
3
-X
4
-
5
-X
6
-X
7
-X
8 -X-Xio-X -Xi 2 -Xi 3 -Xi 4 -XIS-Xi 6
-XI
7 (SEQ ID NO: 43), wherein; 5 Xi is F, Y, H, S, or V;
X
2 is I, or F;
X
3 is Y, S, or W;
X
4 is P, Y, or S; X, is G, S, R, or D; 10 X 6 is D, or G;
X
7 is S, T, G, or R; Xs is E, T, I, or N; X, is T, Y, N, I, K, or H;
X
10 is R, Y, or S; 15 X 11 is Y, N, or S;
X
1 2 is S, P, A, or V;
X
13 is P, S, or D; X1 4 is T, L, or S;
X
1 5 is F, K, or V; 20 X 1 6 is Q, S, or K; and
X
1 7 is G, or is not present; CDR-H3. XI-XrX 3
-X
4
-XS-X-X
7
-X-X
9 -XIO-X-XI2-Xi 3
-X
1 4 -Xi-X 6
-XI
7 -XIB (SEQ ID NO: 44), wherein; X, is V, D, E, S, or C; 25 X 2 is G, R, D, S, K, L, Y, or A;
X
3 is S, G, Y, or R;
X
4 is G, S, Y, N, T, or D;
X
5 is W, S, A, G, Y, or T;
X
6 is Y, G, S, F, W, or N; 30 X 7 is P, S, F, Y, V, G, W, or V; X& is Y, F, D, P, M, I, or N; X, is T, W, D, L, Y, E, P, F, or G;
X
10 is F, D, Y, H, V, Y, or is not present; Xn 1 is D, Y, F, L, or is not present; -3-
X
12 is I, D, Y, or is not present;
X
13 is Y, or is not present;
X
14 is Y, or is not present; XI is G, or is not present; 5 X 16 is M, or is not present;
X
17 is D, or is not present; and
X
8 is V, or is not present; CDR-L1. Xi-X2-X 3
-X
4 -Xs-X,-XrX r-X,-Xio-Xii-XirX-XirXi-XI-Xiy (SEQ ID NO: 45), wherein; 10 X, is R, or K;
X
2 is A, G, or S; X3 is S;
X
4 is E, R, Q, or H;
X
5 is S, I, T, or N; 15 X 6 is I, V, L, or F; X7 is S, G, L, N, or R; Xs is S, G, Y, R, N, H, or D; X, is N, G, Y, R, or S;
X
1 0 is L, Y, S, or D; 20 X 1 , is A, L, N, V, G, or D;
X
1 2 is A, N, E, K, G, or is not present;
X
13 is K, T, N, or is not present;
X
14 is N, Y, T, or is not present;
X
1 5 is Y, L, or is not present; 25 X 16 is L, C, Y, or is not present; and
X
1 , is A, D, or is not present; CDR-L2. XIX 2
-X
3 -XrX4-Xs-Xr-X7 (SEQ ID NO: 46), wherein; X, is T, G, S, W, or E;
X
2 is A, V, T, I, or L; 30 X 3 is S, or F;
X
4 is T, I, N, S, R, or Y;
X
5 is R, or L;
X
6 is A, Q, E, or F; and
X
7 is T, or S; -4- 5 and CDR-L3. Xl-X 2
-X
3
-X
4 -X5-X--X 7 -XS-Xs-Xio (SEQ ID NO: 47), wherein; Xi is Q, or M;
X
2 is Q, H, or Y; 5 X 3 is Y, N, G, S, or R;
X
4 is N, H, Y, D, G, V, L, or ;
X
5 is N, G, I, Y, S, Q, F, or E;
X
6 is W, S, T, L, I, or F;
X
7 is P, L, T, D, or I; 10 X 8 is S, L, P, C, W, I, or F; X, is I, T, S, or is not present; and
X
10 is T, or is not present. 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; 15 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 51-57 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 20 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 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 25 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 30 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; 5 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 10 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; 15 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 D NO.:36; Residues 99-116 of SEQ ID NO.:36; Residues 24-39 of SEQ ID 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 ID NO.:38; Residues 24-35 of 20 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. 25 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; SEQ ID NO: 30; SEQ ID NO: 32; SEQ ID NO: 34; SEQ ID NO: 36; SEQ ID NO: 38; and SEQ 30 ID NO: 40. In another embodiment the binding protein comprises a VL domain. Preferably the VLdomain comprises an amino acid sequence selected from the group consisting 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; -6- 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. In a preferred embodiment the binding protein comprises a VH and a VL domain. More preferably the binding protein comprises a VH domain comprising an amino acid sequence 5 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 and a VL domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 7; SEQ ID NO: 9; SEQ 10 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; and SEQ ID NO: 41. Most 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. 15 In another embodiment the binding protein further comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain; a human IgGI constant domain; a human IgG2 constant domain; a human IgG3 constant domain; a human IgG4 constant domain; a human IgE constant domain and a human IgA constant domain. Preferably the heavy chain immunoglobulin constant region domain is a human IgG1 20 constant domain. Preferably at least one amino acid residue is replaced in the heavy chain constant region domain such that effector functions of the antibody are altered. More preferably the human IgG1 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 light chain 25 immunoglobulin constant domain selected from the group consisting of a human Ig kappa constant domain; and a human Ig lambda constant domain. Preferably the human Ig kappa constant domain comprises amino acid sequence SEQ ID NO.:4 and the human Ig lambda constant domain comprises amino acid sequence SEQ ID NO.:5. In another embodiment the binding protein comprises an Ig constant heavy region having 30 an amino acid sequence selected from the group consisting of: SEQ ID NO:2, and SEQ ID NO: 3; an IG constant light region having an amino acid sequence selected from the group consisting of: SEQ ID NO:4, and SEQ ID NO: 5; an Ig variable heavy region having an amino acid sequence of SEQ ID NO:6; and an Ig variable light region having an amino acid sequence of SEQ ID NO:7 -7- In another embodiment the binding protein comprises an Ig constant heavy region having an amino acid sequence of SEQ ID NO: 3; an IG constant light region having an amino acid sequence of SEQ ID NO:4; an Ig variable heavy region having an amino acid sequence of SEQ ID NO:6; and an Ig variable light region having an amino acid sequence of SEQ ID NO:7. 5 In another embodiment the binding is selected from the group consisting of an immunoglobulin molecule or functional variants thereof known in the art, which variants retain the characteristic binding property of the binding protein. Examples of specific immunoglobulin embodiments include but are not limited to an scFv; a monoclonal antibody; a human antibody; a chimeric antibody; a humanized antibody; a single domain antibody; a Fab fragment; an Fab' 10 fragment; an F(ab')2; an Fv; a disulfide linked Fv, and a bispecific or dual specific antibody. Most preferably the binding protein is a human antibody. Another aspect of the invention provides a neutralizing binding protein comprising any one of the binding proteins disclosed above wherein the neutralizing binding protein is capable of neutralizing IL-18. Preferably the neutralizing binding protein is capable of neutralizing any one 15 of pro-human IL-18; mature-human IL-18 or truncated-human IL-18. In another embodiment the neutralizing binding protein diminishes the ability of IL-18 to bind to its receptor. Preferably the neutralizing binding protein diminishes the ability of pro-human IL-18; mature-human IL-18 or truncated-human IL-18 to bind to its receptor. In another embodiment the neutralizing binding protein is capable of inhibiting one or 20 more of IL-18 biological activities selected from the group consisting of, ThI modulation; Th2 modulation (Nakanishi K., et al (2001) Cytokine and Growth Factor Rev. 12:53-72); Nk modulation; neutrophil modulation; monocyte-macrophage lineage modulation; neutrophil modulation; eosinophil modulation; B-cells modulation; cytokine modulation; chemokine modulation; adhesion molecule modulation; and cell recruitment modulation. 25 In a preferred embodiment the neutralizing binding protein has a dissociation constant (KD) selected from the group consisting of: at most about 10- M; at most about 108 M; at most about 109 M; at most about 1010 M; at most about 10-" M; at most about 1012 M; and at most 10' 1 3 M. In another embodiment the neutralizing binding protein has an on rate selected from the 30 group consisting of: at least about 10 2 M's-; at least about 10 3 Ms; at least about 10 4 M's-; at least about 10 5 's"; and at least about 10 6 Ms". In yet another embodiment the neutralizing binding protein has an off rate selected from the group consisting of: at most about 10 3 s-; at most about 10-4s"; at most about 105s1; and at most about 10-6s-. -8- Another aspect of the invention provides a labeled binding protein comprising any one of the binding proteins disclosed 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 5 biotin. More preferably the radiolabel is 'H, 4 C I 35S, 9Y, 99 Tc, "'In, WI, 131m, "Lu, 'cHo, or 53 Sm. Another aspect of the invention provides a conjugate protein comprising any one of the binding proteins disclosed above wherein said binding protein is conjugated to a therapeutic or cytotoxic agent. Preferably the therapeutic or cytotoxic agent is selected from the group 10 consisting of an anti-metabolite; an alkylating agent; an antibiotic; a growth factor; a cytokine; an anti-angiogenic agent; an anti-mitotic agent; an anthracycline; toxin; and an apoptotic agent. One embodiment pertains to an isolated nucleic acid encoding any one of the binding proteins disclosed above. A further embodiment provides a vector comprising the isolated nucleic acid disclosed above wherein said vector is selected from the group consisting of pcDNA; 15 pTT (Durocher et al., Nucleic Acids Research 2002, Vol 30, No.2); pTT3 (pTT with additional multiple cloning site; pEFBOS (Mizushima, S. and Nagata, S., (1990) Nucleic acids Research Vol 18, No. 17); pBV; pJV; and pBJ. In another embodiment a host cell is transformed with the vector. Preferably the host cell is a prokaryotic cell. More preferably the host cell is E.Coli. In a related embodiment the host 20 cell is an eukaryotic cell. Preferably the eukaryotic cell is selected from the group consisting of protist cell, animal cell, plant cell and fungal cell. More preferably the host cell is a mammalian cell including, but not limited to, CHO and COS; or a fungal cell such as Saccharomyces cerevisiae; or an insect cell such as Sf9. Another aspect of the invention provides a method of producing a binding protein that 25 binds human IL-18, comprising culturing any one of the host cells disclosed 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 30 one of the binding proteins disclosed above, wherein the binding protein exists as a crystal. Preferably the crystal is a carrier- free pharmaceutical controlled release crystal. In one embodiment the binding protein that exists as a crystal has a greater half-life in vivo than the soluble counterpart of the binding protein. In another embodiment the binding protein retains its biological activity after crystallization. -9- One embodiment provides a composition for the release of a binding protein wherein the composition comprises a formulation which in turn comprises a crystallized binding protein as disclosed above and an ingredient; and at least one polymeric carrier. Preferably the polymeric carrier is a polymer selected from one or more of the group consisting of: poly (acrylic acid), poly 5 (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-co-glycolic acid) or PLGA, poly (b-hydroxybutryate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organo)phosphazene], poly (ortho esters), poly (vinyl alcohol), poly (vinylpyrrolidone), maleic anhydride- alkyl vinyl ether copolymers, pluronic polyols, albumin, alginate, cellulose and 10 cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycaminoglycans, sulfated polyeaccharides, blends and copolymers thereof. Preferably the ingredient is selected from the group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl-p- cyclodextrin, methoxypolyethylene glycol and polyethylene glycol. Another embodiment provides a method for treating a mammal comprising the step of administering to the 15 mammal an effective amount of the composition disclosed 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 an IL-18 modulator; 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: 20 NM_000389, NM 002198, NM_002163, NM 006144, NM_006515, NM007185, NM_002288, NM_003661, NM_021958, NM_001335, Hs.382006, NM_020125, NM_007210, NM_021798, NM013324, M11313, D88152, NM_001103, U37519, NM 000697, J03600, NM_014578, S66793, U47054, L19871, M81181, NM_001188, U15460, NM 014417, Z23115, NM_001713, U45878, U37546, U72649, U49187, 25 J03507, U50360 XM_071866, NM_005623, Z32765, Z11697, XM071866, U51096, M83667, D87469, L07765, U66468, X14830, L29217, X15880, NM_001851, M27691, M37435, X13589, X16866, X59131, NM_004393, U73328, L19267, U53445, X68277, U48807, NM_001950, U87269, M57730, X52541, 104076, X63741, L07077, M62831, 30 M60830, U53786, NM_001988, NM_000141, M23668, U60062, NM_000141, U49973, U89995, U27326, A28102, M25667, L34357, U19523, L01406, U03486, X68285, Z18859, D49958, D43772, AC000099, M57731, X53800, M91036, D16583, X64877, X58431, M16937, NM 014468, X92814, L19314, M26665, D10995, L41147, M24283, 35 S81914, J03171, J00219, NM_000619, NM_000585, U31628, X04500, M27492, X01057, M26062, Y00081, Y00787, Z31695, X06256, X57206, U20734, NM_014879, D31762, D42038, NM_005551, NM_014846, X06182, NM_005551, X07730, M13955, M57710, S83362, NM_002314, NM_005569, U49957, U89922, X14008, U59914, D14497, X59727, 40 NM_000429, U43944, X72755, NM_021230, NM 005951, X78710, X70991, M32011, S77763, M58603, S76638, M69043, U91616, D86425, L13740, U44848, U79251, M27288, AF000234, D50640, L20971, L10343, U77735, NM_003579, U17034, AB000584, X63131, D11428, NM_032940, NM_005035, NM_003579, M18255, L01087, D38128, Y10375, -10- D15049, M31166, U59877, NM_003579, U64675, S57153, NM_002903, NG_000013, X75042, M83221, NM_000537, U22314, S59049, U70426, U22377, U38480, L10338, M23178, M69203, NM_.005409, D79206, NM_.005065, NM_004186, J03764, NM_006802, D89077, NM_003037, M91463, 5 D82326, L05568, U96094, X83301, D21267, L31529, M62800, NM_021014, Z35093, NM_005816, L25444, M95787, 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, A1420129, 10 HG2981-HT3127, HG3548-HT3749, HG870-HT870, HG4333-HT4603, HG3111-HT3287, HG4593-HT4998, HG961-HT961, HG1877-HT1917, HG3115-HT3291, HG41 15-HT4385, and HG3925-HT4195. Preferably the modulator is an antagonist. More preferably the modulator is a binding 15 protein or a neutralizing binding protein. The invention also provides a pharmaceutical composition comprising a binding protein or a neutralizing binding protein as disclosed 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 IL-18 activity is detrimental. Preferably the 20 additional agent is selected from the group consisting of: angiogenesis inhibitors (including but not limited to anti-VEGF antibodies or VEGF-trap); kinase inhibitors (including but not limited to KDR and TIE-2 inhibitors); co-stimulation molecule blockers (including but not limited to anti-B7.1, anti-B7.2, CTLA4-Ig, anti-CD20); adhesion molecule blockers (including but not limited to anti-LFA-1 Abs, anti-E/L selectin Abs, small molecule inhibitors); anti-cytokine 25 antibody or functional fragment thereof (including but not limited to anti-IL-12, anti-TNF, anti IL-6/cytokine receptor antibodies); methotrexate; corticosteroids; cyclosporin; rapamycin; FK506; and non-steroidal anti-inflammatory agents. In another aspect, the invention provides a method for inhibiting human IL-18 activity comprising contacting human IL-18 with a binding protein disclosed above such that human IL .30 18 activity is inhibited. In a related aspect the invention provides a method for inhibiting human IL-18 activity in a human subject suffering from a disorder in which IL-18 activity is detrimental, comprising administering to the human subject a binding protein disclosed above such that human IL-18 activity in the human subject is inhibited and treatment is achieved. Preferably the disorder is selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile 35 chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection (including but not limited to bone marrow and solid organ rejection), acute or chronic immune -11disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, 5 sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt's syndrome, adult (acute) respiratory distress syndrome, 10 alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic 15 anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, 20 female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung 25 disease, dermatomyositis/polymyositis associated lung disease, Sjbgren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-i autoimmune 30 hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthbsis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, -12autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, Lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of 5 polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sj6gren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis ,vitiligo, acute liver disease, chronic 10 liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and Th 1 Type mediated diseases, and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and 15 lymphoma). In another aspect the invention provides a method of treating a patient suffering from a disorder in which IL-18 is detrimental comprising the step of administering any one of the binding proteins disclosed above before, concurrent, or after the administration of a second agent, as discussed above. 20 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 binding mature-human IL-18, but does not specifically bind pro-human IL-18. Another aspect of the invention provides a neutralizing binding protein selected from the 25 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 competing with 125-2H antibody for binding human L-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 30 grafted antibody, wherein the neutralizing binding protein is not capable of competing with 125 2H antibody for binding 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 a -13binding protein selected from the group consisting of 2.5(E)mgl antibody and IL-18BP for binding human IL-18. In a preferred embodiment the binding protein is capable of binding mature-human IL-18, but does not specifically bind pro-human IL-18. In yet another embodiment the binding protein is 5 capable of competing with 125-2H antibody for binding human IL-18. In another embodiment the binding protein is not capable of competing with 125-2H antibody for binding human IL-18. In yet another embodiment the binding protein is not capable of competing a binding protein selected from the group consisting of 2.5(E)mgl antibody, and IL-18BP for binding human IL 18. 10 In a preferred embodiment the binding protein comprises a VLdomain comprising an amino acid sequence of SEQ ID NO: 9, and a VH domain comprising an amino acid sequence of SEQ ID NO: 8. In another embodiment the binding protein comprises an Ig constant heavy region having an amino acid sequence selected from the group consisting of: SEQ ID NO:2, and SEQ ID NO: 3; 15 an IG constant light region having an amino acid sequence selected from the group consisting of: SEQ ID NO:4, and SEQ ID NO: 5; an Ig variable heavy region having an amino acid sequence of SEQ ID NO:8; and an Ig variable light region having an amino acid sequence of SEQ ID NO:9. In another embodiment the binding protein comprises an Ig constant heavy region having an amino acid sequence of SEQ ID NO: 3; an IG constant light region having an amino acid 20 sequence of SEQ ID NO:4; an Ig variable heavy region having an amino acid sequence of SEQ ID NO:8; and an Ig variable light region 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 25 antigen-binding portions thereof, that bind thereto. Various 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. Methods of using the antibodies of the invention to detect human IL-i8, to inhibit human IL-18 activity, either in vitro or in vivo, and to regulate gene expression are also encompassed by the 30 invention. This invention also pertains to a truncated IL-18. In related aspects the invention also pertains to nucleic acids, recombinant expression vectors and host cells for making truncated IL 18. Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of -. 14ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, 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 5 "component" encompass both elements and components comprising one unit and elements and components that comprise more than one subunit unless specifically stated otherwise. Generally, nomenclatures used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the 10 art. The methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. Enzymatic reactions and purification techniques are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The 15 nomenclatures used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. 20 That the present invention may be more readily understood, select 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" encompasses native or 25 artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide may 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 native state; is substantially free of other proteins from the same species; 30 is expressed by a cell from a different species; or does not occur in nature. Thus, 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 may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art. -15- The term "recovering" as used herein, refers to the process of rendering a chemical species such as a polypeptide substantially free of naturally associated components by isolation, e.g., using protein purification techniques well known in the art. The term "IL-18" as used herein, refers to the cytokine also known as interferon-gamma 5 inducing factor (IGIF), that is a pro-inflammatory cytokine, that exhibits various functions in addition to an ability to induce interferon gamma. The term "human IL-18" used interchangeably with the term "hIL-18" encompasses polypeptide of SEQ ID NO: 1 and fragments thereof, including but not limited to, pro-human IL-18, mature human IL-18, and any truncated human IL 18 that retains a biological activity of IL-18 as described herein. The term "pro-human IL-18" as 10 used herein, refers to a polypeptide of SEQ ID NO: 1. The term "mature human IL-18" as used herein, refers to residues 37-193 of SEQ ID NO: 1, and the term "truncated human IL-18 as used herein, refers to residues 59-193 of SEQ ID NO: 1. Preferably the IL-18, and fragments thereof, are biologically active. The term "recombinant human IL-18" or "rhIL-18" as used herein, refers to human IL-18 generated in vitro using recombinant DNA techniques. 15 "Biological activity of IL-18" as used herein, refers to all inherent biological properties of the cytokine IL-18. Biological properties of IL-18 include but are not limited to binding IL-18 receptor; promoting maturation and activation of Th1 and Tc 1 cells; promoting production of cytokines such as TNF, IFNy and IL-1P by several cell types; promoting macrophages to release cytokines such as TNF and IFNy, produce NO; promoting FasL expression, cytotoxicity and 20 cytokine release (IFNy) from NK cells; promoting cytokine/chemokine release, respiratory burst, granule release, adhesion molecule expression in Neutrophils; promoting endothelial cells to migrate and thereby promote angiogenesis; promoting GAG release, MMP and NO production in Chondrocytes; promoting COX2 expression in some cells; and reducing cell proliferation in some cells. 25 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 is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A", the 30 presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A" and the antibody, will reduce the amount of labeled A bound to the antibody. The term "antibody", as used herein, broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any -16functional fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule. Such mutant, variant, or derivative anitbody formats are known in the art. Nonlimiting embodiments of which are discussed below. In a full-length antibody, each heavy chain is comprised of a heavy chain variable region 5 (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. 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 one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity 10 determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FRI, CDR1, FR2, CDR2, FR3, CDR3, FR4. The term "antigen-binding portion" of an antibody (or simply "antibody portion"), as 15 used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., 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 embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term "antigen 20 binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHL domains; (ii) a F(ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI 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 25 comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 30 242:423-426; and Huston et al. (1988) Proc. Natl. A cad. Sc. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two -17domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. A cad. Sci. USA 90:6444-6448; Poljak, R.J., et al. (1994) Structure 2:1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody 5 Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5). Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecules, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule 10 (Kipriyanov, S.M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S.M., et al. (1994) Mol. inmunol. 31:1047-1058). Antibody portions, such as Fab and F(ab') 2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole 15 antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein. An "isolated antibody", as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds hIL-18 is substantially free of antibodies that specificallybind 20 antigens other than hIL-18). An isolated antibody that specifically binds hIL-18 may, however, have cross-reactivity to other antigens, such as IL-18 molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. The term "human antibody", as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The 25 human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a 30 mouse, have been grafted onto human framework sequences. The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further in Section II C, below), antibodies isolated from a recombinant, combinatorial human -18antibody library (Hoogenboom H.R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W.E., (2002) Clin. Biochem. 35:425-445; Gavilondo J.V., and Larrick J.W. (2002) BioTechniques 29:128-145; Hoogenboom H., and Chames P. (2000) Immunology Today 21:371 378 ), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human 5 immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295; Kellermann S-A., and Green L.L. (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 that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions 10 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 human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist 15 within the human antibody germline repertoire in vivo. The term "chimeric antibody" refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions. 20 The term "CDR-grafted antibody" refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of 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) has been replaced with human CDR sequences. 25 The term "humanized antibody" refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more "human-like", i.e., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences 30 to replace the corresponding nonhuman CDR sequences. As used herein, the term "hIL-18 neutralizing binding protein" 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 inhibition of a biological activity of hIL-18. Preferably the neutralizing binding protein binds -19hIL-18 and reduces a biologically activity of IL-18 by at least about 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 hIL-18 biological activity. These indicators of hIL-18 biological activity can be assessed by one or more of several standard in vitro or in vivo 5 assays known in the art. The term "epitope" includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor. In certain embodiments, epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional 10 structural characteristics, and/or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody. In certain embodiments, an antibody is said to specifically bind 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 15 that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BlAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see J6nsson, U., et al. (1993) Ann. Biol. Clin. 5_1:19-26; Jnsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnnson, B., et al. 20 (1991) Anal. Biochen. 198:268-277. The term "Kon", as used herein, is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex as is known in the art. The term "Koff", as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex as is known in the art. 25 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 label incorporatedthat provides for the identification of the binding protein. Preferably, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a 30 polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 1 4 C, 35 S, 90 Y, "Tc, 'In, 1 2I, m311, m7Lu, ' 66 Ho, or 1 53 Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels -20- (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates. 5 The term "conjugate binding protein" refers to a binding protein chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent. The term "agent" is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. Preferably the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium 10 bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. The term "crystallized binding protein" as used herein, refers to a polypeptide that 15 exists in the form of a crystal. Crystals are one form of the solid state of matter, which is distinct from other forms such as the amorphous solid state or the liquid crystalline state. Crystals are composed of regular, repeating, three-dimensional arrays of atoms, ions, molecules (e.g., proteins such as antibodies), or molecular assemblies (e.g., antigen/antibody complexes). These three-dimensional arrays are arranged according to specific mathematical relationships 20 that are well-understood in the field. The fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit. Repetition of the asymmetric unit in an arrangement that conforms to a given, well-defined crystallographic symmetry provides the "unit cell" of the crystal. Repetition of the unit cell by regular translations in all three dimensions provides the crystal. See Giege, R. and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a 25 Practical Approach, 2nd ea., pp. 20 1-16, Oxford University Press, New York, New York, (1999)." The term "polynucleotide" as referred to herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxvnucleotides or a modified form of either type of nucleotide. The term includes single and double stranded forms of DNA but preferably is 30 double-stranded DNA. The term "isolated polynucleotide" as used herein shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin, the "isolated polynucleotide": is not associated with all or a portion of a polynucleotide with -21which the "isolated polynucleotide" is found in nature; is operably linked to a polynucleotide that it is not linked to in nature; or does not occur in nature as part of a larger sequence. The term "vector", as used herein, is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a 5 "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal 10 mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, 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 of utility in recombinant DNA techniques are often in the form of 15 plasmids. In the present specification, "plasmid" and "vector" may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. The term "operably linked" refers to a juxtaposition wherein the components described 20 are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence 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. 25 The term "expression control sequence" as used herein refers to polynucleotide sequences which are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance 30 translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The -22term "control sequences" is intended to include components whose presence is essential for expression and processing, and can 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 5 enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such "transformed" cells 10 include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells which 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 intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such 15 terms are intended to refer not only to the particular subject cell, but, to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell" as used herein. Preferably host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life. Preferred eukaryotic 20 cells include protist, fungal, plant and animal cells. Most preferably host cells include but are not limited to the prokaryotic cell line E.Coli; mammalian cell lines CHO and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae. Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and 25 purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory 30 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 -23- "transgene" is a DNA construct, which is stably and operably integrated into the genome of a cell from which a transgenic organism develops, directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic organism. The term "regulate"and "modulate" are used interchangeably, and, as used herein, refers 5 to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of hIL-18). Modulation may be an increase or a decrease 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. 10 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 may cause an increase or decrease 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, 15 which decreases 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, e.g., in WOO1/83525. The term "agonist", as used herein, refers to a modulator that, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the 20 molecule compared to the magnitude of the activity or function observed in the absence of the agonist. Particular agonists of interest may include, but are not limited to, IL-18 polypeptides or polypeptides, nucleic acids, carbohydrates, or any other molecules that bind to hIL-18. The term "antagonist" or "inhibitor", as used herein, refer to a modulator that, when contacted with a molecule of interest causes a decrease in the magnitude of a certain activity or 25 function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist. Particular antagonist of interest include those that block or modulate the biological or immunological activity of h]L- 18. Antagonists and inhibitors of hlL-18 may include, but are not limited to, proteins, nucleic acids, carbohydrates, or any other molecules which bind to hlL-18. 30 The term "sample", as used herein, is used in its broadest sense. A "biological sample", as used herein, includes, but is not limited to, any quantity of a substance from a living thing or formerly 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. -24- The term "competes" as used herein, and as generally known and used by practitioners skilled in the art, refers to the ability of one binding protein to interfere with, or otherwise hinder the binding of a second binding protein to a ligand common to both binding proteins (e.g., IL-18). Assays useful to detemine competition characterisitics of binding proteins are well known in the 5 art. Preferred competition assays are described herein. . Human Antibodies that Bind Human IL-18. One aspect of the present invention provides isolated human antibodies, or antigen 10 binding portions thereof, that bind to IL-18with high affinity, a low off rate and high neutralizing capacity. Preferably, the antibodies, or portions thereof, are isolated antibodies. Preferably, the human antibodies of the invention are neutralizing human anti-IL-18 antibodies. A. Method of making anti IL-18 antibodies Antibodies of the present invention may be made by any of a number of techniques 15 known in the art. A particularly preferred method for generating anti-IL-18 antibodies of the invention include using XENOMOUSE transgenic mice, and using hybridoma and SLAM cellular manipulation techniques (Abgenix, Inc., Fremont, CA) known in the art for preparing antibodies, and using antigens comprising the IL-18 peptide described in Example 3.2, i.e., human IL-18 comprising amino acid sequence of SEQ ID NO. 1 and fragments thereof. 20 In one embodiment of the instant invention, human antibodies are produced by immunizing a non-human animal comprising some, or all, of the human immunoglobulin locus with an IL-18 antigen. In a preferred embodiment, the non-human animal is a XENOMOUSE transgenic mouse, an engineered mouse strain that comprises large fragments of the human immunoglobulin loci and is deficient in mouse antibody production. See, e.g., Green et al. 25 Nature Genetics 7:13-21 (1994) and United States Patents 5,916,771, 5,939,598, 5,985,615, 5,998,209, 6,075,181, 6,091,001, 6,114,598 and 6,130,364. 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, 1996, WO 98/16654, published April 23,1998, WO 98/24893, published June 11, 1998, WO 98/50433, published November 12, 1998, WO 99/4503 1, 30 published September 10, 1999, WO 99/53049, published October 21, 1999, WO 00 09560, published February 24, 2000 and WO 00/037504, published June 29, 2000. The XENOMOUSE transgenic mouse produces an adult-like human repertoire of fully human antibodies, and generates antigen-specific human Mabs. The XENOMOUSE transgenic mouse contains approximately 80% of the human antibody repertoire through introduction of megabase sized, -25germline configuration YAC fragments of the human heavy chain loci and x light chain loci. See Mendez et al., Nature Genetics 15:146-156 (1997), Green and Jakobovits J. Exp. Med. 188:483-495 (1998), the disclosures of which are hereby incorporated by reference. The invention also provides a method for making anti-IL-18 antibodies from non 5 human, non-mouse animals by immunizing non-human transgenic animals that comprise human immunoglobulin loci. One may produce such animals using the methods described immediately above. The methods disclosed in these patents may be modified as described in United States Patent 5,994,619. In a preferred embodiment, the non-human animals may be rats, sheep, pigs, goats, cattle or horses. 10 In another embodiment, the non-human animal comprising human immunoglobulin gene loci are animals that have a "minilocus" of human immunoglobulins. In the minilocus approach, an exogenous Ig locus is mimicked through the inclusion of individual genes from the Ig locus. Thus, one or more VH genes, one or nore DH genes, one or more JH genes, a mu constant region, and a second constant region (preferably a gamma constant region) are 15 formed into a construct for insertion into an animal. This approach is described, inter alia, in U.S. Patent 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, 5,789,215, and 5,643,763, hereby incorporated by reference. An advantage of the minilocus approach is the rapidity with which constructs 20 including portions of the Ig locus can be generated and introduced into animals. However, a potential disadvantage of the minilocus approach is that there may not be sufficient immunoglobulin diversity to support full B-cell development, such that there may be lower antibody production. In order to produce a human anti-IL-18 antibody, a non-human animal comprising some 25 or all of the human immunoglobulin loci is immunized with an IL- 18 antigen and the antibody or the antibody-producing cell is isolated from the animal. The IL-18 antigen may be isolated and/or purified IL- 18 and is preferably a human IL-18. In another embodiment, the IL-18 antigen is a fragment of IL-18, preferably mature IL-18. In another embodiment, the IL-18 antigen is a fragment that comprises at least one epitope of IL-18. 30 Immunization of animals may be done by any method known in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Press, 1990. Methods for immunizing non-human animals such as mice, rats, sheep, goats, pigs, cattle and horses are well known in the art. See, e.g., Harlow and Lane and United States Patent 5,994,619. In a preferred embodiment, the IL-18 antigen is administered with a adjuvant to -26stimulate the immune response. Such adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes). Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local deposit, or they may contain substances that stimulate the host to secrete factors that are chemotactic for 5 macrophages and other components of the immune system. Preferably, if a polypeptide is being administered, the immunization schedule will involve two or more administrations of the polypeptide, spread out over several weeks. Example 2.2.A provides a protocol for immunizing a XENOMOUSE transgenic mouse with human IL-18 in phosphate-buffered saline. 10 B. Production of Antibodies and Antibody-Producing Cell Lines After immunization of an animal with an IL-18 antigen, antibodies and/or antibody producing cells may be obtained from the animal. An anti- IL-18 antibody-containing serum is obtained from the animal by bleeding or sacrificing the animal. The serum may be used as it is obtained from the animal, an immunoglobulin fraction may be obtained from the serum, or the 15 anti- IL-18 antibodies may be purified from the serum. Serum or immunoglobulins obtained in this manner are polyclonal, thus having a heterogeneous array of properties. In another embodiment, antibody-producing immortalized hybridomas may be prepared from the immunized animal. After immunization, the animal is sacrificed and the splenic B cells are fused to immortalized myeloma cells as is well known in the art. See, e.g., Harlow and Lane, 20 supra. In a preferred embodiment, the myeloma cells do not secrete immunoglobulin polypeptides (a non-secretory cell line). After fusion and antibiotic selection, the hybridomas are screened using IL-18, or a portion thereof, or a cell expressing IL-18. In a preferred embodiment, the initial screening is performed using an enzyme-linked immunoassay (ELISA) or a radioimmunoassay (RIA), preferably an ELISA. An example of ELISA screening is provided in 25 WO 00/37504, herein incorporated by reference. Anti- IL-18 antibody-producing hybridomas are selected, cloned and further screened for desirable characteristics, including robust hybridoma growth, high antibody production and desirable antibody characteristics, as discussed further below. Hybridomas may be cultured and expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or 30 in cell culture in vitro. Methods of selecting, cloning and expanding hybridomas are well known to those of ordinary skill in the art. -27- Preferably, the immunized animal is a non-human animal that expresses human immunoglobulin genes and the splenic 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 line is a non-secretory mouse myeloma, such as the 5 myeloma cell line is P3X63Ag8.653 (see, e.g., Example 2.2.B). In one aspect, the invention provides hybridomas that produce human anti- IL-18 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 species such as rats, sheep, pigs, goats, cattle or horses. In another embodiment, the 10 hybridomas are human hybridomas, in which a human non-secretory 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 procedure referred to in the art as the selected lymphocyte antibody method (SLAM), as described in U.S. Patent No. 5,627,052, PCT Publication WO 92/02551 and 15 Babcock, J.S. et al. (1996) Proc. NatI. Acad. Sci. USA 93:7843-7848. In this method, single cells secreting antibodies of interest, e.g., lymphocytes derived from any one of the immunized animals described in Section I (A), are screened using an antigen-specific hemolytic plaque assay, wherein the antigen IL-18, or a fragment thereof, is coupled to sheep red blood cells using a linker, such as biotin, and used to identify single cells that secrete antibodies with specificity 20 for IL-18. Following identification of antibody-secreting cells of interest, heavy- and light-chain variable region cDNAs are rescued from the cells by reverse transcriptase-PCR and these variable regions can then be expressed, in the context of appropriate immunoglobulin constant regions (e.g., human constant regions), in mammalian host cells, such as COS or CHO cells. The host cells transfected with the amplified immunoglobulin sequences, derived from in vivo selected 25 lymphocytes, can then undergo further analysis and selection in vitro, for example by panning the transfected cells to isolate cells expressing antibodies to IL-18. The amplified immunoglobulin sequences further can be manipulated in vitro, such as by in vitro affinity maturation methods such as those described in PCT Publication WO 97/29131 and PCT Publication WO 00/56772. In vitro methods also can be used to make the antibodies of the invention, wherein an 30 antibody library is screened to identify an antibody having the desired binding specificity. Methods for such screening of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al. U.S. Patent No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; -28- Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al. PCT Publication No. WO 92/01047; Garrard et al. PCT Publication No. WO 92/09690; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; McCafferty et al., Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 5 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982, US patent application publication 20030186374, and PCT Publication No. WO 97/29131, the contents of each of which are incorporated herein by reference. 10 The recombinant antibody library may be from a subject immunized with IL-18, or a portion of IL-18. Alternatively, the recombinant antibody library may be from a naYve subject, i.e., one who has not been immunized with IL-18, such as a human antibody library from a human subject who has not been immunized with human IL-18. Antibodies of the invention are selected by screening the recombinant antibody library with the peptide comprising human IL-18 (e.g., a 15 peptide corresponding to a portion of hIL-18) to thereby select those antibodies that recognize IL 18. Methods for conducting such screening and selection are well known in the art, such as described in the references in the preceding paragraph. To select antibodies of the invention having particular binding affinities for hIL-18, such as those that dissociate from human IL-I 8 with a particular k,,,r rate constant, the art-known method of surface plasmon resonance can be 20 used to select antibodies having the desired kf rate constant. To select antibodies of the invention having a particular neutralizing activity for hIL-18, such as those with a particular an
IC
50 , standard methods known in the art for assessing the inhibition of hIL-18 activity may be used. In one aspect, the invention pertains to an isolated antibody, or an antigen-binding 25 portion thereof, that binds human 11-18. 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 ID NO: 7 and VH with amino acid sequence of SEQ ID NO: 6. Most preferrably, the 2.5(E) antibody binds 30 human IL-18 with a Kdof less than 5x101 0 M (see Example 2.2.F). Preferrably, anti-IL-18 antibodies of the present invention, such as the 2.5(E) antibody and related antibodies, exhibit a capacity to reduce or to neutralize IL-18 activity, e.g.,as assessed by any one of several in vitro and in vivo assays known in the art (e.g., see Example 3.2.F). For example, these antibodies neutralize IL-18-induced production of human interferon gamma in -29- KG-1 cells with ICso values in the range of at least about 10- M, about 101 M, or about 10-10 M. Further these antibodies also neutralize IL-18-induced production of human interferon gamma in the whole blood cells with IC 50 values in the range of at least about 10'8 M, about 109 M, or about 10.1 M. 5 In a particularly preferred embodiment the anti-IL- 18 antibody 2.5(E) binds to human IL 18 in various forms, including pro-IL-18, mature IL-18 and truncated IL-18. The antibody 2.5(E) does not specifically bind to other cytokines, such as IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, ]L-13, IL-15, IL-16, IL-17, IL-21, TNF, LT (lymphotoxin), LTcal P2, and LTct2$l. However, the antibody 2.5(E) does exhibit cross reactivity to IL-18 from other species. 10 For example, the antibody neutralizes the activity of IL-18 from cynomolgus monkey (IC50 for cyno IL-18= 9.1E X 10~"; See Example 2.2.J1). In one aspect, the invention pertains to 2.5(E) antibodies and functional antibody portions, 2.5(E) -related antibodies and functional antibody portions, and other human antibodies and functional antibody portions with equivalent properties to 2.5(E), such as high affinity 15 binding to IL-18 with low dissociation kinetics and high neutralizing capacity. In prefered 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 with a koff rate constant of about 0. Is- or less, as determined by surface plasmon resonance, or which inhibits human IL-18 activity with an IC 50 of about 1 x 10- 6 M or less. Alternatively, the antibody, 20 or an antigen-binding portion thereof, may dissociate from human IL-18 with a kog rate constant of about 1 x 104sdor less, as determined by surface plasmon resonance, or may inhibit human IL 18 activity with an IC 5 0 of about I x 10 7 M or less. Alternatively, the antibody, or an antigen binding portion thereof, may dissociate from human IL- 18 with a k.f rate constant of about 1 x 10-3S-1 or less, as determined by surface plasmon resonance, or may inhibit human IL-i 8 activity 25 with an IC 50 of about .1 x 10- 7 M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-18 with a kof rate constant of about 1 x 10-4s 1 or less, as determined by surface plasmon resonance, or may inhibit human IL-18 activity with an IC 5 o of about 1 x 10-M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-18 with a kof rate constant of about 1 x 10-s- or less, as determined by 30 surface plasmon resonance, or may inhibit human IL-18 activity with an IC 50 of about 1 x 101 0 M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-18 with a kff rate constant of about 1 x 0- 5 s~'or less, as determined by surface plasmon resonance, or may inhibit human IL-18 activity with an IC 50 of about 1 x 10"M or less. -30- In still 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 5 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 heavy chain variable region (VH) comprising an amino acid sequence 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 10 NO: 36; SEQ ID NO: 38; or SEQ ID NO: 40. In certain embodiments, the antibody comprises a heavy chain constant region, such as an IgGI, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region. Preferably, the heavy chain constant region is an IgGI heavy chain constant region or an IgG4 heavy chain constant region. Furthermore, the antibody can comprise a light chain constant region, either a kappa light chain 15 constant region or a lambda light chain constant region. Preferably, the antibody comprises a kappa light chain constant region. Alternatively, the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment. Replacements of amino acid residues in the Fc portion to alter antibody effector function are known in the art (Winter, et al. US PAT NOS 5,648,260; 5624821). The Fc portion of an 20 antibody mediates several important effector functions e.g. cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/ clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives. Certain human IgG isotypes, particularly IgGI and IgG3, mediate 25 ADCC and CDC via binding to FcyRs and complement Clq, respectively. Neonatal Fc receptors (FcRn) are the critical components determining the circulating half-life of antibodies. In still another embodiment at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered. 30 One embodiment provides a labeled binding protein wherein an antibody or antibody portion of the invention is derivatized 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 linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, -31such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can'mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag). 5 Useful detectable agents with which an antibody or antibody portion of the invention may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase 10 and the like. When an antibody is derivatized 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 agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or 15 streptavidin binding. Another embodiment of the invention provides a crystallized binding protein. Preferably the invention relates to crystals of whole anti-IL-18 antibodies and fragments thereof as disclosed herein, and formulations and compositions comprising such crystals. In one embodiment the crystallized binding protein has a greater half-life in vivo than the soluble counterpart of the 20 binding protein. In another embodiment the binding protein retains biological activity after crystallization. Crystallized binding protein of the invention may be produced according methods known in the art and as disclosed in WO 02072636, incorporated herein by reference. (Also see Example 2.2.M) 25 Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues. Nascent in vivo protein production may undergo further processing, known as post-translational modification. In particular, sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation. The resulting proteins bearing covalently linked oligosaccharide side 30 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 may produce different glycosylation enzymes (eg., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl -32residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid. Preferably the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human. 5 It is known to those skilled in the art that differing protein glycosylation may result in differing protein characteristics. For instance, the efficacy of a therapeutic protein produced in a microorganism host, such as yeast, and glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line. Such glycoproteins may also be immunogenic in humans and show reduced half-life in 10 vivo after administration. Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream. Other adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity. Accordingly, a practitioner may prefer a therapeutic protein with a 15 specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal. Expressing glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using 20 techniques known in the art a practitioner may generate antibodies or antigen-binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins (glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S patent applications 20040018590 and 25 20020137134). Further, it will be appreciated by one skilled in the art that a protein of interest may be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that member host cells of the library produce the protein of interest with variant glycosylation patterns. A practitioner may then select and isolate the protein of interest with 30 particular novel glycosylation patterns. Preferably, the protein having a particularly selected novel glycosylation pattern exhibits improved or altered biological properties. -33- C. Production of recombinant IL-18 antibodies Antibodies of the present invention may be produced by any of a number of techniques known in the art. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is(are) transfected into a host cell by standard techniques. 5 The various forms of the term "transfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells is 10 preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active 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 Urlaub and 15 Chasin, (1980) Proc. Nati. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e:g., as described in R.J. Kaufman and P.A. Sharp (1982) Mol. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells. When 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 expression of the antibody in the host cells or, more 20 preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods. Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are 25 within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules 30 are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods. -34- In a preferred system for recombinant expression of an antibody, or antigen-binding portion thereof, of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy 5 and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is 10 recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium. Still further the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant antibody of the invention is 15 synthesized. The method can further comprise isolating the recombinant antibody from the culture medium. Table 1 is a list of amino acid sequences of VH and VL regions of preferred anti-hIL-18 antibodies of the invention. In the VH region, the naturally occuring amino acid in position 1 of the amino terminus (N-terminus) is either Glutamate (E) or Glutamine (Q). However, to generate 20 recombinant protein with homogeneous N-termini during large-scale production of protein comprising VH region, Glutamate (E) is preferred in position 1 of the N-terminus. Table 1 List of Amino Acid Sequences of VH and VL regions Protein Sequence Protein region Sequence Identifier 12345678901234567890 EVQLVQSGTEVKKPGESLKI SCKGSGYTVTSYWIGWVRQM PGKGLEWMGFIYPGDSETRY VH 2.5(E) SEQ ID NO.:6 SPTFQGQVTISADKSFNTAF LQWSSLKASDTAMYYCARVG SGWYPYTFDIWGQGTMVTVS S VH 2.5 CDR-H1 Residues 31-35 of SEQ ID NO.:6 SYWIG VH 2.5 CDR-H2 Residues 50-66 of SEQ ID NO.:6 FIYPGDSETRYSPTFQG VH 2.5 CDR-H3 Residues 99-110 of SEQ ID NO.:6 VGSGWYPYTFDI EIVMTQSPATLSVSPGERAT LSCRASESISSNLAWYQQKP GQAPRLFIYTASTRATDIPA VL 2.(E) SEQ ID NO.:7 RFSGSGSGTEFTLTISSLQS EDFAVYYCQQYNNWPSITFG _QGTRLEIKR -35- 36 Protein Sequence Protein region Sequence Identifier 12345678901234567890 VL 2.5 CDR-L1 Residues 24-34 of SEQ ID NO.:7 RASESISSNLA VL 2.5 CDR-L2 Residues 50-56 of SEQ ID NO.:7 TASTRAT VL 2.5 CDR-L3 Residues 89-98 of SEQ ID NO.:7 QQYNNWPSIT QVQLQESGPGLVTPSQTLSL TCTVSGGSISSGGHYWTWIR QHPGKGLEWIGYIYYSGSTY VH 2.13 SEQ ID NO.:8 YNPSLKSRLTISVDTSKNQF SLKLSSVAAADTAVYYCARD RGGSGSYWDYWGQGTLVTVS iS VH 2.13 CDR-H1 Residues 31-37 of SEQ ID NO.:8 SGGHYWT VH 2.13 CDR-H2 Residues 52-67 of SEQ ID NO.:8 YIYYSGSTYYNPSLKS VH 2.13 CDR-H3 Residues 100-110 of SEQ ID NO.:8 DRGGSGSYWDY EIVLTQSPGTLSLSPGERAT LSCRGSRSVSSGVLAWYQQK PGQAPRLLIYGVSIRATGIP VL 2.13 SEQ ID NO.:9 DRFSGSGSGTDFTLTISRLE PEDFAVYYCQQYHGSPLTFG . ._____ _GGTKVEIKR VL 2.13 CDR-L1 Residues 24-35 of SBQ ID NO.:9 RGSRSVSSGYLA VL 2.13 CDR-L2, Residues 51-57 of SEQ ID NO.:9 GVSIRAT VL 2.13 CDR-L3 Residues 90- 98 of SEQ ID NO.:9 QQYHGSPLT QVQLQESGPGLVKPSETLSL TCTVSGGSIRNYYWSWIRQP PGKGLEWVGYIYSSGSTNYN VH 2.3 SEQ ID NO.:10 PSLKSRVTISVDTSKNQFSL KLSSVTAADTAVYYCARDRG GASFFDYWGQGTLVTVSS VHI 2.3 CDR-H1 Residues 31-35 of SEQ ID NO.:10 NYYWS VH 2.3CDR-H2 Residues 50-65 of SEQ ID NO.:10 YIYSSGSTNYNPSLKS VII 2.3 CDR-H3 Residues 98-107 of SEQ ID NO.: 10 DRGGASFFDY DIQMTQSPSSLSASIGDRVT ITCRASQIIGGYLNWYQQRP GKAPKFLIYSTSILQSGVPS VL 2.3 SEQ ID NO.:11 RFSGSGSGTDFTLTISSLQP EDFATYYCQQTYITPPTFGP GTKVDIKR VL 2.3 CDR-L1 Residues 24-34 of SEQ ID NO.: 11 RASQIIGGYLN VL 2.3 CDR-L2 Residues 50-56 of SEQ ID NO.:11 STSILQS VL 2.3 CDR-L3 Residues 89-97 of SEQ ID NO.: 11 QQTYITPPT IRVQLQESGPGLVKPSQTLSL TCTVSGGSINSGDYYWSWIRI QHPGKGLEWIGHISYRGTTY VH 215 SEQ ID NO.:12 YNPSLKSRVTISVDTSKNQF SLKL SSVTAADTAVYCCARD _ __ RGGGFFDLWGRGTLVTVSS VH 215 CDR-H1 Residues 31-37 of SEQ ID NO.:12 SGDYYWS VH 215 CDR-H2 Residues 52-67 of SEQ ID NO.:12 HISYRGTTYYNPSLKS VH 215 CDR-H3 Residues 100-108 of SEQ ID NO.:12 DRGGGFFDL Protein Sequence Protein region Sequence Identifier 12345678901234567890 EIVLTQSPGTLSLSPGERAT LSCRASRSLSSGYLAWYQQK PGQAPRLLIYGASIRATGIP VL 215 SEQ I) NO.13 DRFSGSGSATDFTLTISRLE PEDFAVYYCQQYNYSPLTFG GGTRVEINR VL 215 CDR-L1 Residues 24-35 of SEQ ID NO.:13 RASRSLSSGYLA VL 215 CDR-L2 Residues 51-57 of SEQ ID NO.:13 GASIRAT VL 215 CDR-L3 Residues 90-98 of SEQ ID NO.:13 QQYNYSPLT EVQLVESGGGSVQPRGSLRL SCAASGFTFSSYSMNWVRQA PGKGLEWVSYFSSSGGIIYY VH 231 SEQ ID NO.:14 ADSVKGRFTISRDNAKNSLY LQMNSLRDEDTAVYYCARDD SSGYYPYFFDYWGQGTLVTV SS VH 231 CDR-H1 Residues 31-35 of SEQ ID NO.:14 SYSMN VH 231 CDR-H2 Residues 50-66 of SEQ ID NO.: 14 YFSSSGGIIYYADSVKG VH 231 CDR-H3 Residues 99-111 of SEQ ID NO.: 14 DDSSGYYPYFFDY DIVMTQSPDSLAVSLGERAT INCKSSQTVLYRSNNKNYLA WYQQKSGQPPKLLIYWASTR VL 231 SEQ ID NO.:15 ESGVPDRFSGSGSGTDFTLT ISSLQAEDVAVYYCQQYYST PLTFGGGTKVEIKR VL 231 CDR-L1 Residues 24-40 of SEQ ID NO.: 15 KSSQTVLYRSNNKNYLA VL 231 CDR-L2 Residues 56-62 of SEQ ID NO.:15 WASTRES VL 231 CDR-L3 Residues 95-103 of SEQ ID NO.: 15 QQYYSTPLT QLQLQESGPGLVKPSETLSL TCTVSGGSISSRVYYWGWIR QPPGKGLEWIGSIYYSGSTY VH 251 SEQ ID NO.:16 YNPSLKSRVTISVDASKNQF SLKLSSVTAADTAIYYCARE DSSAWVFEHWGQGTLVTVSS VH 251 CDR-H1 Residues 31-37 of SEQ ID NO.:16 SRVYYWG VH 251 CDR-H2 Residues 52-67 of SEQ ID NO.:16 SIYYSGSTYYNPSLKS VH 251 CDR-H3 Residues 100-109 of SEQ ID EDSSAWVFEH NO.:16 EIVLTQSPDTLSLSPGERAT LSCRASHILSRNYLAWYQQK PGQAPRLLMYGISIRATGIP VL 251 SEQ ID NO.: 17 DRFSGSGSGADFTLTINRLE PEDFAVYYCQHYDNSLCSFG QGTKLEVKR VL 251 CDR-L1 Residues 24-35 of SEQ ID NO.:17 RASHILSRNYLA VL 251 CDR-L2 Residues 51-57 of SEQ ID NO.:17 GISIRAT VL 251 CDR-L3 Residues 90-98 of SEQ ID NO.:17 QHYDNSLCS -37- Protein Sequence Protein region Sequence Identifier 12345678901234567890 QVQLVESGGGVVQPGRSLRL SCAASGFTFRNYGLHWVRQA PGKGLEWVAVIWYDGSNKYY VH 268 SEQ ID NO.:18 ADSVKGRFTISRDNSKNTLY LQMNSLRAEDTAVYYCARES YYYYGMDVWGQGTTVTVSS VH 268 CDR-H1 Residues 31-35 of SEQ ID NO.:1 8 NYGLH VH 268 CDR-H2 Residues 20-36 of SEQ ID NO.:18 VIWYDGSNKYYADSVKG VH 268 CDR-H3 Residues 99-108 of SEQ ID NO.: 18 ESYYYYGMDV EIVMTQSPATLSVSPGERAT LSCRASQSFNSNLVWYQQKP GQAPRLLIYGASTRATGIPA SEQ ID NO.:19 RFSGSGSGTEFTLTISSLQS VL 268 EDFAVYYCQQYNNWTWTFGQ GTKVEIKR VL 268 CDR-L1 Residues 24-34 of SEQ ID NO.:19 RASQSFNSNLV VL 268 CDR-L2 Residues 50-56 of SEQ ID NO.:19 GASTRAT VL 268 CDR-L3 Residues 89-97 of SEQ ID NO.: 19 QQYNNWTWT QVQLQESGPGLVKPSQTLSL TCTVSGGSINSGDYYWSWIR QHPGKGLEWIGHISYRGTTY VH 336 SEQ ID NO.:20 YNPSLKSRVTISVDTSKNQF SLKLSSVTAADTAVYCCARD RGGGFFDLWGRGTLVTVSS VH 336 CDR-H1 Residues 31-35 of SEQ ID NO.:20 SGDYYWS VH 336 CDR-H2 Residues 52-67 of SEQ ID NO.:20 HISYRGTTYYNPSLKS VH 336 CDR-H3 Residues 100-108 of SEQ ID NO.:20 DRGGGFFDL EIVLTQSPGTLSLSPGERAT LSCRASQSVSSGYLAWYQQK PGQAPRLLIYGASIRATGIP VL 336 SEQ ID NO.:21 DRFSGSGSATDFTLTISRLE PEDFAVYYCQQYGYSPLTFG GGTRVEINR VL 336 CDR-LI Residues 24-35 of SEQ ID NO.:21 RASQSVSSGYLA VL 336 CDR-L2 Residues 51-57 of SEQ ID NO.:21 GASIRAT VL 336 CDR-L3 Residues 90-98 of SEQ ID NO.:21 QQYGYSPLT QVQLVESGGGVVQPGRSLRL SCAASGFTFSHYGMHWVRQA PGKGLEWVAVISYDGRNKYY VH 351 SEQ ID NO.:22 VDSVKGRFTISRDNSKNTLY LQMNSLRAEDTAVFYCAREK GGSGWPPFYYYYGMDVWGQG TTVTVSS VH 351 CDR-Hl Residues 31-35 of SEQ ID NO.:22 HYGMH VH 351 CDR-H2 Residues 50-66 of SEQ ID NO.:22 VISYDGRNKYYVDSVKG VH 351 CDR-H3 Residues 99-116 of SEQ ID NO.:22 EKGGSGWPPFYYYYGMDV -38- Protein Sequence Protein region Sequence Identifier 12345678901234567890 DIVMTQTPLSLSVTPGQPAS ISCKSSQNLLYSDGETYLCW YLQKPGQPPQLLIYEVSNRF SGVPERFSGSGSGTDFTLKI VL 351 SEQ ID NO.:23 SRVEAEDVGIYYCMQNVQLP LTFGGGTRVEIKR VL 351 CDR-L1 Residues 24-39 of SEQ ID NO.:23 KSSQNLLYSDGETYLC VL 351 CDR-L2 Residues 55-61 of SEQ ID NO.:23 EVSNRFS VL 351 CDR-L3 Residues 94-102 of SEQ ID NO.:23 MQNVQLPLT QTQLQESGPGLVKPSETLSL TCTVSGGSISSRVYYWGWIR QPPGKGLEWIGSIYYSGSTY VH 413 SEQ ID NO.:24 YSPSLKSRVTISVDTSKNQF SLKLSSVTAADTAIYYCARE DSSAWVFEHWGQGTLVTVSS VH 413 CDR-HI Residues 31-37 of SEQ ID NO.:24 SRVYYWG VH 413 CDR-H2 Residues 52-67 of SEQ ID NO.:24 SIYYSGSTYYSPSLKS VH 413 CDR-H3 Residues 100-109 of SEQ ID NO.:24 EDSSAWVFEH EIVLTQSPDTLSLSPGERAT LSCRASQILSRNYLAWYQQK PGQAPRLLIYGISIRATGIP VL 413 SEQ ID NO.:25 DRFSGSGSGADFTLTINRLE PEDFAVYYCQHYDNSLCSFG QGTKLEVKR VL 413 CDR-L1 Residues 24-35 of SEQ ID NO.:25 RASQILSRNYLA VL 413 CDR-L2 Residues 51-57 of SEQ ID NO.:25 GISIRAT VL 413 CDR-L3 Residues 90-98 of SEQ ID NO.:25 QHYDNSLCS QLQLQESGPGLVKPSETLSL TCTVSGGSIDSRIYYWGWIR QPPGKGLEWIGSIYYRGSTY VH 435 SEQ ID NO.:26 YNPSLKSRVTISVDTPKNQF SLKLNSVTAADTAVYYCARE DSSAWVFDYWGQGTLATVSS VH 435 CDR-HI Residues 31-37 of SEQ ID NO.:26 SRIYYWG VH 435 CDR-H2 Residues 52-67 of SEQ ID NO.:26 SIYYRGSTYYNPSLKS VH 435 CDR-H3 Residues 100-109 of SEQ ID NO.:26 EDSSAWVFDY EIVLTQSPGTLSLSPGERAT LSCRASQSVRNNYLNWYQQK PGQAPRLLIYGAFSRATGIP VL 435 SEQ ID NO.:27 DRFSGSGSGTDFTLTISSLE PEDFVVYYCQQYGNSIDSFG QGTKLEINR VL 435 CDR-L1 Residues 24-35 of SEQ ID NO.:27 RASQSVRNNYLN VL 435 CDR-L2 Residues 51-57 of SEQ ID NO.:27 GAFSRAT VL 435 CDR-L3 Residues 90-98 of SEQ ID NO.:27 QYGNSIDS -39- Protein Sequence Protein region Sequence Identifier 123456789012345.67890 QVQLQESGPGLVKPSQTLSL TCTVSGGSINSGDYYWSYIR QHPGKGLEWIGHISYRGTTY VH 444 SEQ ID NO.:28 YNPSLKSRVTISVDTSKNQF SLKLSSVTAADTAVYCCARD RGGGFFDLWGRGTLVTVSS VH 444 CDR-H1 Residues 31-37 of SEQ ID NO.:28 SGDYYWS VH 444 CDR-H2 Residues 52-67 of SEQ ID NO.:28 HISYRGTTYYNPSLKS VH 444 CDR-H3 Residues 100-108 of SEQ I) NO.:28 DRGGGFFDL EIVLTQSPGTLSLSPGERAT LSCRASQSVSSGYLAWYQRK PGQAPRLLIYGTSIRATGIP VL 444 SEQ ED NO.:29 DRFSGSGSATDFTLSISRLG PEDFAVYYCQQYGYSPLTFG GGTRVEINR VL 444 CDR-LI Residues 24-35 of SEQ ID NO.:29 RASQSVSSGYLA VL 444 CDR-L2 Residues 51-57 of SEQ ID NO.:29 GTSIRAT VL 444 CDR-L3 Residues 90-98 of SEQ ID NO.:29 QQYGYSPLT QVQLQESGPGLVKPSQTLSL TCTVSGGSISSGGHYWSWIR QHPGKGLEWIGYIYYSGSTH VH 478 SEQ ID NO.:30 YNPSLKSRVTISVDTSKNQF SLKLRSVSAADTAGYYCASL YNGNGYFDLWGRGTLVTVSS VH 478 CDR-H1 Residues 31-37 of SEQ ID NO.:30 SGGHYWS VH 478 CDR-H2 Residues 52-67 of SEQ ID NO.:30 YIYYSGSTHYNPSLKS VH 478 CDR-H3 Residues 99-109 of SEQ ID NO.:30 SLYNGNGYFDL EIVLTQSPGTLSLSPGERAT LSCRASQSISSGYLAWYQQK PGQAPRLIIYGVSRRATGIP VL 478 SEQ ID NO.:31 DRFSGSGSGADFTLTISRLD PEDFVVYYCQQYGFSPLTFG GGTKVEIKR VL 478 CDR-L1 Residues 24-35 of SEQ ID NO.:31 RASQSISSGYLA VL 478 CDR-L2 Residues 51-57 of SEQ ID NO.:31 GVSRRAT VL 478 CDR-L3 Residues 90-98 of SEQ ID NO.:31 QQYGFSPLT QLQLQESGPGLVKPSETLSL TCTVSGGSISRSYDYWGWIR QPPGKGLEWIGSIYYRGSTY VH 521 SEQ ID NO.:32 YNPSLKSRVTISVDTSKNQF SLKLSSVTAADTAVYYCARE YSTTWSIDYWGQGTLVTVSS VH 521 CDR-H1 Residues 31-37 of SEQ ID NO.:32 RSYDYWG VH 521 CDR-H2 Residues 52-67 of SEQ ID NO.:32 SIYYRGSTYYNPSLKS VH 521 CDR-H3 Residues 100-109 of SEQ ID EYSTTWSIDY NO.:32 -40- Protein Sequence Protein region Sequence Identifier 12345678901234567890 ENVLTQSPGTLSLSPGERAT LSCRASQSIRNNYLAWYQQK PGQAPRLLIHGASSRATGIP VL 521 SEQ ID NO.:33 DRFGGSGSGTDFTLTISRLE PEDFAVYFCQQYGNSIITFG PGTKVDVNR VL 521 CDR-L1 Residues 24-35 of SEQ ID NO.:33 RASQSIRNNYLA VL 521 CDR-L2 Residues 51-57 of SEQ ID NO.:33 GASSRAT VL 521 CDR-L3 Residues 90-98 of SEQ ID NO.:33 QQYGNSIIT QVQLQESGPGLVKPSQTLSL TFCTVSGGSINSGGYYWSWIR QHPGKGLEWIGHISYRGTTY VH 550 SEQ ID NO.:34 SNPSLKSRVTISVDTSKNQF SLKLSSVTAADTAVYYCARD RGGGFFDLWGRGTLVTVSS VH 550 CDR-H1 Residues 31-37 of SEQ ID NO.:34 SGGYYWS VH 550 CDR-H2 Residues 52-67 of SEQ ID NO.:34 HISYRGTTYSNPSLKS VH 550 CDR-H3 Residues 100-108 of SEQ ID NO.:34 DRGGGFFDL EIVLTQSPGTLSLSPGERAT LSCRASQSVNSGYLAWYQQK PGQAPRLLIYGVSIRATDIP VL 550 SEQ ID NO.:35 DRFSGSGSATDFTLTISRLE PEDFAVYYCQQYGFSPLTFG GGTRVEINR VL 550 CDR-L1 Residues 24-35 of SEQ ID NO.:35 RASQSVNSGYLA VL 550 CDR-L2 Residues 51-57 of SEQ ID NO.:35 GVSIRAT VL 550 CDR-L3 Residues 90-98 of SEQ ID NO.:35 QQYGFSPLT QVQLVESGGGVVQPGRSLRL SCAASGFTFSHCGMHWVRQA PGKGLEWVAVISYDGSNKYY VH 581 SEQ ID NO.:36 ADSVKGRFTISRDNSKNTLY LQMNSLRAEDTAVYYCAKDH GGSGSPPFYYYYGMDVWGQG TTVTVSS VH 581 CDR-H1 Residues 31-35 of SEQ ID NO.:36 HCGMH VH 581 CDR-H2 Residues 50-66 of SEQ ID NO.:36 VISYDGSNKYYADSVKG VH 581 CDR-H3 Residues 99-116 of SEQ ID NO.:36 DHGGSGSPPFYYYYGMDV DILMTQTPLSLSVTPGQPAS ISCKSSQSLLHGDGKTYLYW YLQKPGQPPQFLIQELSNRF VL 581 SEQ ID NO.:37 SGVPDRFSGSGSGTDFTLKI SRXEAEDVGVYYCMQSLQLP LTFGGGTKVQIKR VL 581 CDR-L1 Residues 24-39 of SEQ ID NO.:37 KSSQSLLHGDGKTYLY VL 581 CDR-L2 Residues 55-61 of SEQ ID NO.:37 ELSNRFS VL 581 CDR-L3 Residues 94-102 of SEQ ID NO.:37 MQSLQLPLT -41- Protein Sequence Protein region Sequence Identifier 12345678901234567890 QVQLVESGGGVVQPGRSLRL SCAASGFTFSYYGMHWVRQA PGKGLEWVAVIWYDGRNKYY VH 7.5 SEQ ID NO.:38 ADSVKGRVTISRDNSKKTLY LQMNSLRAEDTAVYYCAREG GYYYGMDVWGQGTTVTVSS VH 7.5 CDR-H1 Residues 31-35 of SEQ ID NO.:38 YYGMH VH 7.5 CDR-H2 Residues 50-66 of SEQ ID NO.:38 VIWYDGRNKYYADSVKG VH 7.5 CDR-H3 Residues 99-108 of SEQ ID NO.:38 EGGYYYGMDV EILLTQSPGTLSLSPGERAT LSCRASQNVSSSYLAWYQQN PGQAPRLLIYGASSRATGIP VL 7.5 SEQ ID NO.:39 DRFSGSGSGTDFTLTISRLE PEDFEVYYCQQSGSSLFTFG PGTKVDIKR VL 7.5 CDR-L1 Residues 24-35 of SEQ ID NO.:39 RASQNVSSSYLA VL 7.5 CDR-L2 Residues 51-57 of SEQ ID NO.:39 GASSRAT VL 7.5 CDR-L3 Residues 90-98 of SEQ ID NO.:39 QQSGSSLFT 2VQLQESGPGLVKPSQTLSL TCTVSGGSIRSGDHYWTWIR QHPGKGLEWIGHIYYSGSTY VH 2.11 SEQ ID NO.:40 YNPSLKSRLTISIDTSKNQF SLKLSSVTAADTAVYYCARD YGGNGYFDYWGQGTLVTVSS VH 2.11 CDR-H1 Residues 31-37 of SEQ ID NO.:40 SGDHYWT VH 2.11 CDR-H2 Residues 52-67 of SEQ ID NO.:40 HIYYSGSTYYNPSLKS VH 2.11 CDR-H3 Residues 97-109 of SEQ ID NO.:40 CARDYGGNGYFDY DIVMTQTPLSLPVTPGEPAS ISCRSSQSLLDSDDGNTYLD WYLQKPGQSPQLLIYTLSYR VL 2.11 SEQ ID NO.:41 ASGVPDRFSGSGSGTDFTLN ISRVEAEDVGVYYCMQRIEF PITFGQGTRLEIKR VL 2.11 CDR-L1 Residues 24-40 of SEQ ID NO.:41 RSSQSLLDSDDGNTYLD VL 2.11 CDR-L2 Residues 56-62 of SEQ ID NO.:41 TLSYRAS VL 2.11 CDR-L3 Residues 95-103 of SEQ ID NO.:41 MQRIEFPIT The foregoing isolated anti-IL-18 antibody CDR sequences establish a novel family of IL-18 binding proteins, isolated in accordance with this invention, and comprising polypeptides that include the CDR sequences listed in Table 2 below. To generate and to select CDR's of the 5 invention having preferred IL-18 binding and/or neutralizing activity, standard methods known in the art for generating binding proteins of the present invention and assessing the IL-18 binding and/or neutralizing characteristics of those binding protein may be used, including but not limited to those specifically described herein. -42- Table 2: Consensus IL-18 CDR affinity ligands (alternative residues are listed below each amino acid position; - indicates residue may be absent). CDR Sequence Consensus region Identifier Sequence CDR-H1 SEQ ID X 1
X
2
X
3
X
4
X
5 XG X 7 NO.:42 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-H2 SEQ ID X 1
X
2
X
3
X
4
X
5
X
6 X7 X8 X 9
X
10 Xu X12 X 13
X
14 Xis X 16
X
17 NO.:43 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-H3 SEQ ID X 1
X
2
X
3
X
4
X
5
X
6
X
7
X
8
X
9 Xio X11 X 12
X
13
X
14
X
15
X
16
X
1 7 Xi NO.:44 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 L D T N G I E V Y W N P Y A V F G CDR-L1 SEQ ID X 1
X
2
X
3
X
4
X
5
X
6
X
7
X
8
X
9 X1o X 11
X
1 2
X
13
X
1 4
X
15 Xi 6
X
1 7 NO.:45 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 L C D H N F N R R D V E N T Y R N S G K H D G D CDR-L2 SEQ ID XI X 2
X
3
X
4
X
5
X
6
X
7 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-L3 SEQ ID X 1
X
2
X
3
X
4 Xs X 6
X
7 Xs X 9
X
1 NO.:47 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 L F F I E -43- D. Uses of Anti-IL-18 Antibodies Given their ability to bind to human IL-18, the 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 5 serum or plasma), using a conventional immunoassay, such as an enzyme linked immunosorbent assays (ELISA), an radioimmunoassay (RIA) or tissue immunohistochemistry. The invention provides a method for detecting 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 human IL- 18 or unbound antibody (or antibody portion), 10 to thereby detect human IL-18 in the biological sample. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the 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, 0-galactosidase, or acetylcholinesterase; examples 15 of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3 H. 1 4 C 35s, "Y, 9 Tc, 1"In, 1251, 1311, Lu, 16 6 Ho, or 1 5 3 Sm. 20 Alternative to labeling the antibody, human IL-I8 can be assayed in biological fluids by a competition immunoassay utilizing rhIL-18 standards labeled with a detectable substance and an unlabeled anti- human IL-18 antibody. In this assay, the biological sample, the labeled rhIL-18 standards and the anti- human IL-18 antibody are combined and the amount of labeled rhIL-18 standard bound to the unlabeled antibody is determined. The amount of human IL-18 in the 25 biological sample is inversely proportional to the amount of labeled rhIL-18 standard bound to the anti-IL-18 antibody. The antibodies and antibody portions of the invention preferably are capable of neutralizing human IL-18 activity both in vitro and in vivo. Accordingly, such antibodies and antibody portions of the invention can be used to inhibit hIL-18 activity, e.g., in a cell culture 30 containing hIL-18, in human subjects or in other mammalian subjects having IL-18 with which an antibody of the invention cross-reacts. In one embodiment, the invention provides a method for inhibiting IL-18 activity comprising contacting IL-18 with an antibody or antibody portion of the invention such that IL-18 activity is inhibited. Preferably, the IL-18 is human IL-18. For example, in a cell culture containing, or suspected of containing hIL-18, an antibody or antibody -44portion of the invention can be added to the culture medium to inhibit hIL-18 activity in the culture. In another embodiment, the invention provides a method for reducing IL-18 activity in a subject, advantageously from a subject suffering from a disease or disorder in which IL-18 5 activity is detrimental. The invention provides methods for reducing IL-18 activity 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 such that IL-18 activity in the subject is reduced. Preferably, the IL-18 is human IL-18 and the subject is a human subject. Alternatively, the subject can be a mammal expressing an IL-18 to which an antibody of the invention is capable of 10 binding. Still further the subject can be a mammal into which hIL-18 has been introduced (e.g., by administration of hIL-18 or by expression of an hIL-18 transgene). An antibody of the invention can be administered to a human subject for therapeutic purposes. Moreover, 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 15 disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration). As used herein, the term "a disorder in which IL-18 activity is detrimental" is intended to include diseases and other disorders in which the presence of IL-18 in a subject suffering from 20 the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which IL-18 activity is detrimental is a disorder in which reduction of IL-18 activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of IL-18 in a 25 biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of IL-18 in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using 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 pertaining to pharmaceutical compositions of the antibodies of the invention. 30 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 -45one embodiment, the pharmaceutical composition further comprises at least one additional therapeutic agent for treating a disorder in which IL-18 activity is detrimental. The antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the 5 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 solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate 10 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. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or 15 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 composed of either a liquid or lyophilized dosage form in a flint or 20 amber vial, ampule or pre-filled syringe. The buffer can be L-histidine (1-50 mM), optimally 5 10mM, at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers 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, 25 principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trenhalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 2-4%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-Methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). 30 Additional surfactants include but are not limited to polysorbate 20 and BRU surfactants. The compositions of this invention may be 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 intended mode of administration and -46therapeutic application. Typical preferred 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 5 administered by intravenous infusion or injection. In another preferred embodiment, the antibody is administered by intramuscular or subcutaneous injection. 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 to high drug concentration. Sterile 10 injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the 15 case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of 20 surfactants. Prolonged absorption of injectable compositions can be brought about by including, in the composition, an agent that delays absorption, for example, monostearate salts and gelatin. 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 25 be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may 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, 30 polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. -47- In certain embodiments, an antibody or antibody portion of the invention may be orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic 5 administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. 10 Supplementary active compounds can also be incorporated into the compositions. In certain embodiments, an antibody or antibody portion of the invention is coformulated with and/or coadministered with one or more additional therapeutic agents that are useful for treating disorders in which IL-18 activity is detrimental. For example, an anti-hIL-18 antibody or antibody portion of the invention may be coformulated and/or coadministered with one or more 15 additional antibodies that bind other targets (e.g., antibodies that bind other cytokines or that bind cell surface molecules). Furthermore, one or more antibodies of the invention may be used in combination with two or more of the foregoing therapeutic agents. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. 20 In certain embodiments, an antibody to IL-18 or fragment thereof is linked to a half-life extending vehicle known in the art. Such vehicles include, but are not limited to, the Fc domain, polyethylene glycol, and dextran. Such vehicles are described, e.g., in U.S. Application Serial No. 09/428,082 and published PCT Application No. WO 99/25044, which are hereby incorporated by reference for any purpose. 25 Interleukin 18 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, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, 30 thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic -48active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, 5 sporadic, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoinunune bullous 10 disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, 15 Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypoganmmaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis 20 associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjugren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, fibrosis, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic 25 pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with 30 organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalnia, pulmonary hypertension secondary to connective tissue -49disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjurgren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism 5 (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and Th1 Type mediated diseases, acute 10 and chronic pain (different forms of pain), and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma). The human antibodies, and 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 15 uveitis. 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, mellitus and psoriasis. An antibody, or antibody portion, of the invention also can be administered with one or 20 more additional therapeutic agents useful in the treatment of autoimmune and inflammatory diseases. Antibodies of the invention, or antigen binding portions thereof can be used alone or in combination to treat such diseases. It should be understood that the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with an additional agent, 25 e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention. The additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition. 30 It should further be understood that the combinations which are to be included within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limited. The combinations, which are part of this invention, can be the antibodies of the present invention and at least one additional agent selected from the lists below. The combination can also include more than one additional agent, -50e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function. Preferred combinations are non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS which include drugs like ibuprofen. Other preferred combinations are corticosteroids 5 including prednisolone; the well known side-effects of steroid use can be reduced or even eliminated by tapering the steroid dose required when treating patients in combination with the anti-IL-18 antibodies of this invention. Non-limiting examples of therapeutic agents for rheumatoid arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); 10 antibodies to 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, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or 15 their ligands including CD154 (gp39 or CD40L). Preferred combinations of therapeutic agents may interfere at different points in the autoimmune and subsequent inflammatory cascade; preferred examples include TNF antagonists like chimeric, humanized or human TNF antibodies, D2E7, (PCT Publication No. WO 97/29131), CA2 (Remicadem), CDP 571, and soluble p55 or p75 TNF receptors, derivatives, thereof, 20 (p75TNFRlgG (Enbrelm) or p55TNFR1gG (Lenercept), and also TNFt converting enzyme (TACE) inhibitors; similarly IL-1 inhibitors (Interleukin-1-converting enzyme inhibitors, IL-1RA etc.) may be effective for the same reason. Other preferred combinations include Interleukin 11. Yet another preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with 1L-18 function; especially preferred are IL-12 25 antagonists including IL-12 antibodies or soluble IL-12 receptors, or IL-12 binding proteins. It has been shown that 1L-12 and IL-18 have overlapping but distinct functions and a combination of antagonists to both may be most effective. Yet another preferred combination are non depleting anti-CD4 inhibitors. Yet other preferred combinations include antagonists of the co stimulatory pathway CD80 (B7.1) or CD86 (B7.2) including antibodies, soluble receptors or 30 antagonistic ligands. The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 -51adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, 5 antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFax or IL-I (e.g. IRAK, NIK, IKK , p38 or MAP kinase inhibitors), IL-1B converting enzyme inhibitors, TNFax converting enzyme (TACE) inhibitors, T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble 10 cytokine receptors and derivatives thereof (e.g. soluble p55 or p7 5 TNF receptors and the derivatives p75TNFRIgG (Enbrel" and p55TNFRIgG (Lenercept)), sIL-1RI, sIL-1RII, sIL-6R), antiinflammatory cytokines (e.g. IL-4, IL-10, IL-i1, IL-13 and TGFP), celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium 15 thiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate/apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone hcl, hydrocodone bitartrate/apap, diclofenac sodium/misoprostol, fentanyl, anakinra, human recombinant, tramadol hcl, salsalate, sulindac, cyanocobalamin/fa/pyridoxine, acetaminophen, alendronate sodium, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, glucosamine 20 sulf/chondroitin, amitriptyline hcl, sulfadiazine, oxycodone hcl/acetaminophen, olopatadine hcl, misoprostol, naproxen sodium, omeprazole, cyclophosphamide, rituximab, IL-1 TRAP, MRA, CTLA4-IG,,IL-l8 BP, anti-IL-12, Anti-IL15, BIRB-796, SCIO-469, VX-702, AMG-548, VX 740, Roflumilast, IC-485, CDC-801, and Mesopram. Preferred combinations include methotrexate or leflunomide and in moderate or severe rheumatoid arthritis cases, cyclosporine. 25 Non-limiting examples of therapeutic agents for inflammatory bowel disease with which an antibody, or antibody portion, of the invention can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane inhibitors; IL-1 receptor 30 antagonists; anti-IL-1p monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-15, IL 16, 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, -52- CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands. The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, 5 adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFa or IL-I (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), IL-IP converting enzyme inhibitors, TNFa converting enzyme inhibitors, T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme 10 inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-1RI, sIL-IRII, sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-ll, IL 13 and TGF$). 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, for example, 15 anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/2913 1; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL) and p55TNFRIgG (LENERCEPT)) inhibitors and PDE4 inhibitors. Antibodies of the invention, or antigen binding portions thereof, can be combined with corticosteroids, for example, budenoside and dexamethasone. Antibodies of the invention or antigen binding portions thereof, may also be 20 combined with agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine, and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-1, for example, IL-13 converting enzyme inhibitors and IL-ira. Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6-mercaptopurines. Antibodies of the invention, or antigen binding portions thereof, 25 can be combined with IL-i1. Antibodies of the invention, or antigen binding portions 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, 30 cholestyramine/sucrose, ciprofloxacin hydrochloride, hyoscyamine sulfate, meperidine hydrochloride, midazolam hydrochloride, oxycodone hcl/acetaminophen, promethazine hydrochloride, sodium phosphate, sulfamethoxazole/trimethoprim, celecoxib, polycarbophil, propoxyphene napsylate, hydrocortisone, multivitamins, balsalazide disodium, codeine -53phosphate/apap, colesevelam hcl, cyanocobalamiin, 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: 5 corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-aminopyridine; tizanidine; interferon-1la (AVONEX; Biogen); interferon-olb (BETASERON; Chiron/Berlex); interferon a-n3) (Interferon Sciences/Fujimoto), interferon-a (Alfa Wassermann/J&J), interferon $1A-IF (Serono/Inhale Therapeutics), Peginterferon a 2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1; COPAXONE; Teva 10 Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; antibodies to or antagonists of other human cytokines or growth factors and their receptors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-23, IL-15, IL-16, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, 15 CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands. The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents 20 which interfere with signalling by proinflammatory cytokines such as TNFa or IL-1 (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), IL-15 converting enzyme inhibitors, TACE inhibitors, T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-IRI, sIL-lRII, sIL 25 6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-13 and TGFO). Preferred examples of therapeutic agents for multiple sclerosis in which the antibody or antigen binding portion thereof can be combined to include interferon-$, for example, IFNp Ia and IFN$Ib; copaxone, corticosteroids, caspase inhibitors , for example inhibitors of caspase-1, IL-1 inhibitors, TNF inhibitors, and antibodies to CD40 ligand and CD80. 30 The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as alemtuzumab, dronabinol, Unimed,daclizumab,mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunokine NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2778, calagualine, CPI-1189, LEM (liposome encapsulated mitoxantrone), THC.CBD (cannabinoid agonist) MBP -54- 8298, mesopram (PDE4 inhibitor), MNA-7 15, anti-IL-6 receptor antibody, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-R1, talampanel, teriflunomide,TGF-beta2, tiplimotide, VLA-4 antagonists (for example, TR-14035, VLA4 Ultrahaler, Antegran-ELAN/Biogen), interferon gamma antagonists, IL-4 agonists. 5 Non-limiting examples of therapeutic agents 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, amlodipine besylate, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, verapamil hcl, digoxin, propranolol 10 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 fumarate. Non-limiting examples of therapeutic agents for Ankylosing Spondylitis with which an 15 antibody, or antibody portion, of the invention can be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, Sulfasalazine, Methotrexate, azathioprine, minocyclin, prednisone, etanercept, infliximab. Non-limiting examples of therapeutic agents for Asthma with which an antibody, or 20 antibody portion, of the invention can be combined include the following: albuterol, salmeterol/fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, levalbuterol hcl, albuterol sulfate/ipratropium, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pirbuterol acetate, prednisolone, theophylline anhydrous, methylprednisolone 25 sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin trihydrate, flunisolide, allergy injection, cromolyn sodium, fexofenadine hydrochloride, flunisolide/menthol, amoxicillin/clavulanate, levofloxacin, inhaler assist device, guaifenesin, dexamethasone sodium phosphate, moxifloxacin hcl, doxycycline hyclate, guaifenesin/d-methorphan, p-ephedrine/cod/chlorphenir, gatifloxacin, cetirizine 30 hydrochloride, mometasone furoate, salmeterol xinafoate, benzonatate, cephalexin, pe/hydrocodone/chlorphenir, cetirizine hcl/pseudoephed, phenylephrine/cod/promethazine, codeine/promethazine, cefprozil, dexamethasone, guaifenesin/pseudoephedrine, chlorpheniramine/hydrocodone, nedocromil sodium, terbutaline sulfate, epinephrine, methylprednisolone, metaproterenol sulfate. -55- 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, theophylline anhydrous, methylprednisolone sodium 5 succinate, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, levalbuterol hcl, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin/clavulanate, flunisolide/menthol, chlorpheniramine/hydrocodone, metaproterenol sulfate, methylprednisolone, mometasone furoate, p-ephedrine/cod/chlorphenir, pirbuterol 10 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 conl, Interferon-alpha-nl, Pegylated interferon-alpha-2a, 15 Pegylated interferon-alpha-2b, ribavirin, Peginterferon alfa-2b + ribavirin, Ursodeoxycholic Acid, Glycyrrhizic Acid, Thymalfasin, Maxamine, VX-497 and any compounds that are used to treat HCV through intervention with the following targets:HCV polymerase, HCV protease, HCV helicase, HCV IRES (internal ribosome entry site). Non-limiting examples of therapeutic agents for Idiopathic Pulmonary. Fibrosis with 20 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 sod succ, lorazepam, furosemide, lisinopril, nitroglycerin, spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, oxycodone hcl, potassium chloride, 25 triamcinolone acetonide, tacrolimus anhydrous, calcium, interferon-alpha, methotrexate, mycophenolate mofetil, Interferon-gamma-10. 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, 30 carvedilol, atenolol, morphine sulfate, metoprolol succinate, warfarin sodium, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril hel/mag carb, bumetanide, alteplase, enalaprilat, arniodarone hydrochloride, tirofiban hcl m-hydrate, diltiazem hydrochloride, captopril, irbesartan, valsartan, propranolol hydrochloride, fosinopril sodium, lidocaine -56hydrochloride, eptifibatide, cefazolin sodium, atropine sulfate, aminocaproic acid, spironolactone, interferon, sotalol hydrochloride, potassium chloride, docusate sodium, dobutamine hcl, alprazolam, pravastatin sodium, atorvastatin calcium, midazolam hydrochloride, meperidine hydrochloride, isosorbide dinitrate, epinephrine, dopamine hydrochloride, 5 bivalirudin, rosuvastatin, ezetimibe/simvastatin, avasimibe, cariporide. Non-limiting examples of therapeutic agents 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, tazarotene, methotrexate, fluocinonide, betamethasone diprop augmented, fluocinolone acetonide, acitretin, tar shampoo, 10 betamethasone valerate, mometasone furoate, ketoconazole, pramoxine/fluocinolone, hydrocortisone valerate, flurandrenolide, urea, betamethasone, clobetasol propionate/emoll, fluticasone propionate, azithromycin, hydrocortisone, moisturizing formula, folic acid, desonide, pimecrolimus, coal tar, diflorasone diacetate, etanercept folate, lactic acid, methoxsalen, hc/bismuth subgal/znox/resor, methylprednisolone acetate, prednisone, sunscreen, halcinonide, 15 salicylic acid, anthralin, clocortolone pivalate, coal extract, coal tar/salicylic acid, coal tar/salicylic acid/sulfur, desoximetasone, diazepam, emollient, fluocinonide/emollient, mineral oil/castor oil/na lact, mineral oil/peanut oil, petroleum/isopropyl myristate, psoralen, salicylic acid, soap/tribromsalan, thimerosal/boric acid, celecoxib, infliximab, cyclosporine, alefacept, efalizumab, tacrolimus, pimecrolimus, PUVA, UVB, sulfasalazine. 20 Non-limiting examples of therapeutic 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 acetate, indomethacin, hydroxychloroquine sulfate, prednisone, sulindac, betamethasone diprop augmented, infliximab, methotrexate, folate, triamcinolone acetonide, 25 diclofenac, dimethylsulfoxide, piroxicam, diclofenac sodium, ketoprofen, meloxicam, methylprednisolone, nabumetone, tolmetin sodium, calcipotriene, cyclosporine, diclofenac sodium/misoprostol, fluocinonide, glucosamine sulfate, gold sodium thiomalate, hydrocodone bitartrate/apap, ibuprofen, risedronate sodium, sulfadiazine, thioguanine, valdecoxib, alefacept, efalizumab. 30 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, tacrolimus, ABT-578, acetaminophen. Non-limiting examples of therapeutic agents for Sciatica with which an antibody, or antibody portion, of the invention can be combined include the following: hydrocodone -57bitartrate/apap, rofecoxib, cyclobenzaprine hcl, methylprednisolone, naproxen, ibuprofen, oxycodone hcl/acetaminophen, celecoxib, valdecoxib, methylprednisolone acetate, prednisone, codeine phosphate/apap, tramadol hcl/acetaminophen, metaxalone, meloxicam, methocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapentin, dexamethasone, carisoprodol, ketorolac 5 tromethamine, indomethacin, acetaminophen, diazepam, nabumetone, oxycodone hcl, tizanidine hcl, diclofenac sodium/misoprostol, propoxyphene napsylate/apap, asa/oxycod/oxycodone ter, ibuprofen/hydrocodone bit, tramadol hcl, etodolac, propoxyphene hcl, amitriptyline hcl, carisoprodol/codeine phos/asa, morphine sulfate, multivitamins, naproxen sodium, orphenadrine citrate, temazepam. 10 Preferred examples of therapeutic agents for SLE (Lupus) in which an antibody or an antigen binding portion can be combined include the following: NSAIDS, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexamethasone; Cytotoxics, for example, 15 azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; inhibitors of PDE4 or purine synthesis inhibitor, for example Cellcept. Antibodies of the invention or antigen binding portions thereof, may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-1, for example, caspase inhibitors like IL-10 converting 20 enzyme inhibitors and IL-Ira. Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-IgG or anti-B7 family antibodies, anti-PD-1 family antibodies. Antibodies of the invention, or antigen binding portions thereof, can be combined with IL-11 or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg 25 antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to B-cell surface molecules. Antibodies of the invention or antigen binding portion thereof may also be used with LP 394 (abetimus), agents that deplete or inactivate B-cells, for example, Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS antibody), TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 30 (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. A "therapeutically effective amount" refers to an amount effective, at dosages and -58for 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 skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the 5 individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically 10 effective amount will be less than the therapeutically effective amount. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous 15 to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms 20 of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. An exemplary, non-limiting range for a therapeutically or prophylactically effective 25 amount of an antibody or antibody portion of the invention is 0.1-20 mg/kg, more preferably 1-10 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and 30 that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. -59- 11. IL-18 responsive genes IL-18 is expressed in macrophages, dendritic cells, Kupffer cells, microglia, epithelial cells, keratinocytes, intestinal epithelial cells, chondrocytes, synovial fibroblasts and osteoblasts, as well as within the adrenal cortex and pituitary gland. In some cells, such as human monocytes 5 and dendritic cells, expression is constitutive, whereas in other cells it must be induced de novo. Apart from interferon gamma expression, little is known about other genes induced by IL-18 alone or in concert with other cytokines. One embodiment of the invention provides a method for regulating gene expression of a gene of interest comprising the steps of providing IL-18 or an IL-18 modulator; and contacting 10 IL-18 or the modulator to a cell wherein the gene of interest is selected from the group consisting of the genes presented in the following table. Table 3 IL-18 Responsive Genes Genbank ID Gene Name Unigene Comment NM_000389 p2 cyclin-dependent kinase inhibitor 1A (p21, Cip1) NM_002198 IRF1 interferon regulatory factor I NM_002163 ICSBP1 interferon consensus sequence binding protein 1 NM 006144 GZMA granzyme A NM-006515 SETMAR SET domain and mariner transposase fusion gene NM_007185 TNRC4 trinucleotide repeat containing 4 NM 002288 LAIR2 leukocyte-associated Ig-like receptor 2 NM 003661 APOL1 apolipoprotein L, 1 NM-021958 HLX1 H2.0-like homeo box 1 (Drosophila) NM_001335 CTSW cathepsin W (lymphopain) Hs.382006 FCGR1B FcRI b form (AA 1-344) NM 020125 BLAME leucine aminopeptidase 3 NM..007210 GALNT6 UDP-N-acetyl-alpha-D-galactosamine NM_021798 IL21R interleukin 21 receptor NM_013324 CISH cytokine inducible SH2-containing protein M11313 A2M alpha-2-macroglobulin D88152 ACATN acetyl-Coenzyme A transporter NM_001103 ACTN2 actinin, alpha 2 U37519 ALDH8 aldehyde dehydrogenase 8 NM_000697 ALOX12 arachidonate 12-lipoxygenase J03600 ALOX5 arachidonate 5-lipoxygenase NM_014578 ARHD ras homolog gene family, member S66793 ARR3 arrestin 3, retinal (X-arrestin) U47054 ART3 ADP-ribosyltransferase 3 L19871 ATF3 activating transcription factor 3 M81181 ATP1B2 ATPase, Na+/K+ transporting NM_001188 BAKI BCL2-antagonist/killer 1 U15460 BATF basic leucine zipper transcription factor, ATF-like NM_014417 BBC3 Bcl-2 binding component 3 Z23115 BCL2L1 BCL2-like 1 NM 001713 BHMT betaine-homocysteine methyltransferase -60- U45878 BIRC3 baculoviral LAP repeat-containing 3 U37546 BIRC3 baculoviral IAP repeat-containing 3 U72649 BTG2 BTG family, member 2 U49187 C60RF32 chromosome 6 open reading frame 32 J03507 C7 complement component 7 U50360 CAMK2G CaM kinase II gamma XM 071866 CAT56 CAT56 protein NM_005623 CCL8 Z32765 CD36 CD36 antigen (collagen type I/ TSP receptor) HG2981- CD44 CD44 antigen HT3127 Z1 1697 CD83 CD83 antigen XM 071866 CDR2 cerebellar degeneration-related protein (62kD) U51096 CDX2 caudal type homeo box transcription factor 2 M83667 CEBPD CCAAT/enhancer binding protein (C/EBP), delta D87469 CELSR2 cadherin, EGF LAG seven-pass G-type receptor 2 L07765 CES 1 carboxylesterase 1 U66468 CGR11 cell growth regulatory with EF-hand domain X14830 CHRNB1 cholinergic receptor, nicotinic, beta polypeptide 1 L29217 CLK3 CDC-like kinase 3 X15880 COL6A1 collagen, type VI, alpha 1 NM 001851 COL9A1 collagen, type IX, alpha 1 M27691 CREB 1 cAMP responsive element binding protein I M37435 CSFI colony stimulating factor 1 (macrophage) HG3548- CUTL1 cut (CCAAT displacement protein) HT3749 X13589 CYP19 cytochrome P450, subfamily XIX X16866 CYP2D7AP cytochrome P450, subfamily IID X59131 D13S106E highly charged protein NM_004393 DAGI dystroglycan 1 U73328 DLX4 distal-less homeobox 4 L19267 DMWD dystrophia myotonica, WD repeat motif U53445 DOC1 downregulated in ovarian cancer 1 X68277 DUSP1 dual specificity phosphatase 1 U48807 DUSP4 dual specificity-phosphatase 4 NM_001950 E2F4 E2F transcription factor 4, p107/p130-binding U87269 E4F1 E4F transcription factor 1 M57730 EFNA1 ephrin-Al X52541 EGR1 early growth response I J04076 EGR2 early growth response 2 (Krox-20 homolog) X63741 EGR3 early growth response 3 L07077 EHHADH enoyl-Coenzyme A M62831 ETR101 immediate early protein M60830 EVI2B ecotropic viral integration site 2B U53786 EVPL envoplakin NM_001988 EVPL envoplakin NM_000141 FCGBP Fc fragment of IgG binding protein M23668 FDX1 ferredoxin 1 U60062 FEZI fasciculation & elongation protein zeta 1 (zygin I) -61- NM_000141 FGFR2 fibroblast growth factor receptor 2 U49973 FLJ10803 hypothetical protein FLJ10803 U89995 FOXE1 forkhead box El (thyroid transcription factor 2) U27326 FUT3 fucosyltransferase 3 A28102 GABRA3 gamma-aminobutyric acid (GABA) receptor M25667 GAP43 growth associated protein 43 L34357 GATA4 GATA-binding protein 4 U19523 GCH1 GTP cyclohydrolase 1 L01406 GHRHR growth hormone releasing hormone receptor U03486 GJA5 gap junction protein, alpha 5, 40kD (connexin 40) X68285 GK glycerol kinase Z18859 GNAT2 guanine nucleotide binding protein (G protein) HG870-HT870 GOLGA3 golgi autoantigen, golgin subfamily a, 3 D49958 GPM6A glycoprotein M6A D43772 GRB7 growth factor receptor-bound protein 7 AC000099 GRM8 glutamate receptor, metabotropic 8 M57731 GRO2 GRO2 oncogene X53800 GRO3 GRO3 oncogene M91036 HBG2 hemoglobin, gamma G D16583 HDC histidine decarboxylase X64877 HFL3 H factor (complement)-like 3 X58431 HOXB6 homeo box B6 M16937 HOXB7 homeo box B7 NM_014468 HPX42B haemopoietic progenitor homeobox X92814 HREV107 similar to rat HREV 107 L19314 HRY hairy (Drosophila)-homolog M26665 HTN3 histatin 3 D10995 HTRIB 5-hydroxytryptamine (serotonin) receptor 1B L41 147 HTR6 5-hydroxytryptarnine (serotonin) receptor 6 M24283 ICAMI intercellular adhesion molecule I (CD54) S81914 IER3 immediate early response 3 J03171 IFNAR1 interferon (alpha, beta and omega) receptor I J00219 IFNG interferon, gamma NM_000619 IFNG interferon, gamma NM-000585 IL15 interleukin 15 U31628 IL15RA interleukin 15 receptor, alpha X04500 IL1B interleukin 1, beta M27492 IL1R1 interleukin 1 receptor, type I X01057 IL2RA interleukin 2 receptor, alpha M26062 IL2RB interleukin 2 receptor, beta Y00081 ]L6 interleukin 6 (interferon, beta 2) Y00787 IL8 interleukin 8 Z31695 INPP5A inositol polyphosphate-5-phosphatase, 40kD X06256 ITGA5 integrin, alpha 5 X57206 1TPKB inositol 1,4,5-trisphosphate 3-kinase B U20734 JUNB jun B proto-oncogene NM 014879 KIAAOOO1 putative G protein coupled receptor for UDP-glucose D31762 KIAA0057 TRAM-like protein D42038 KIAA0087 KIAA0087 gene product -62- NM 005551 KIAA0133 KLAA0133 gene product NM 014846 KIAA0196 KIAA0196 gene product X06182 KIT v-kit oncogene homolog NM 005551 KLK2 kallikrein 2, prostatic X07730 KLK3 kallilrein 3, (prostate specific antigen) M13955 KRT7 keratin 7 M57710 LGALS3 lectin, galactoside-binding, soluble, 3 (galectin 3) S83362 LIFR leukemia inhibitory factor receptor NM 002314 LIMK1 LIM domain kinase 1 NM 005569 LIMK2 LIM domain kinase 2 U49957 LPP LIM domain-containing U89922 LTB lymphotoxin beta (TNF superfamily, member 3) X14008 LYZ lysozyme (renal amyloidosis) U59914 MADH6 MAD ) homolog 6 D14497 MAP3K8 mitogen-activated protein kinase kinase kinase 8 X59727 MAPK4 mitogen-activated protein kinase 4 NM_000429 MATlA methionine adenosyltransferase I, alpha HG1877- MBP myelin basic protein HT1917 , HG3115- MBP myelin basic protein HT3291 U43944 ME1 malic enzyme 1, NADP(+)-dependent, cytosolic X72755 MIG monokine induced by gamma interferon NM_021230 MLL3 myeloid/lymphoid or mixed-lineage leukemia3 NM 005951 MT1H metallothionein 1H X78710 MTF1 metal-regulatory transcription factor 1 X70991 NAB2 NGFI-A binding protein 2 (ERG 1 bp 2) M32011 NCF2 neutrophil cytosolic factor 2 S77763 NFE2 nuclear factor (erythroid-derived 2), 45kD M58603 NFKB1 nuclear factor kappa B (p105) S76638 NFKB2 nuclear factor kappa B M69043 NFKBIA nuclear factor kappa B U91616 NFKBIE nuclear factor kappa B D86425 NID2 nidogen 2 L13740 NR4A1 nuclear receptor subfamily 4, group A, member 1 U44848 NRF1 nuclear respiratory factor 1 U79251 OPCML opioid-binding protein/cell adhesion molecule-like HG4115- OR1B3P olfactory receptor HT4385 M27288 OSM oncostatin M AF000234 P2RX4 purinergic receptor P2X D50640 PDE3B phosphodiesterase 3B, cGMP-inhibited L20971 PDE4B phosphodiesterase 4B, cAMP-specific L10343 P13 protease inhibitor 3, skin-derived (SKALP) U77735 PIM2 pim-2 oncogene NM_003579 PIP5K2A phosphatidylinositol-4-phosphate 5-kinase U17034 PLA2R1 phospholipase A2 receptor 1, l80kD AB000584 PLAB prostate differentiation factor X63131 PML promyelocytic leukemia -63- D11428 PMP22 peripheral myelin protein 22 NM 032940 POLR2C polymerase (RNA) I polypeptide NM 005035 POLRMT polymerase (RNA) mitochondrial (DNA directed) NM003579 POU2F2 POU domain, class 2, transcription factor 2 M18255 PRKCB1 protein kinase C, beta 1 L01087 PRKCQ protein kinase C, theta D38128 PTGIR prostaglandin 12 (prostacyclin) receptor (IP) Y10375 PTPNS1 tyrosine phosphatase, non-receptor substrate 1 D15049 PTPRH protein tyrosine phosphatase, receptor type, H M31166 PTX3 pentaxin-related gene, U59877 RAB31 RAB31, member RAS oncogene family NM_003579 RAD54L RAD54 (S.cerevisiae)-like U64675 RANBP2L1 RAN binding protein 2-like 1 S57153 RBBP1 retinoblastoma-binding protein I NM_002903 RCV1 recoverin NG_000013 RDBP RD RNA-binding protein X75042 REL v-rel M83221 RELB v-rel NM_000537 REN renin U22314 REST REl-silencing transcription factor S59049 RGS1 regulator of G-protein signalling I U70426 RGS 16 regulator of G-protein signalling 16 U22377 RLF rearranged L-myc fusion sequence U38480 RXRG retinoid X receptor, gamma L10338 SCN1B sodium channel polypeptide M23178 SCYA3 small inducible cytokine A3 M69203 SCYA4 small inducible cytokine A4 NM 005409 SCYB 11 small inducible cytokine subfamily B: CXC1 1 D79206 SDC4 syndecan 4 (amphiglycan, ryudocan) NM_005065 SELIL sel-1 (suppressor of lin-12, C.elegans)-like NM_004186 SEMA3F semaphorin 3F J03764 SERPINE1 nexin, plasminogen activator inhibitor type 1 NM_006802 SF3A3 splicing factor 3a, subunit 3, 60kD HG3925- SFTPA2 surfactant, pulmonary-associated protein A2 HT4195 D89077 SLA Src-like-adapter NM_003037 SLAM signaling lymphocytic activation molecule M91463 SLC2A4 solute carrier family 2 glucose transporter D82326 SLC3A1 solute carrier family 3 L05568 SLC6A4 solute carrier family 6 (serotonin), U96094 SLN sarcolipin X83301 SMA3 SMA3 D21267 SNAP25 synaptosomal-associated protein, 25kD L31529 SNTB1 syntrophin, dystrophin-associated protein Al, HG961-HT961 SOSI son of sevenless (Drosophila) homolog 1 M62800 SSA1 (52kD, ribonucleoprotein autoantigen SS-A/Ro) NM 021014 SSX3 synovial sarcoma, X breakpoint 3 Z35093 SURFI surfeit 1 NM_005816 TACTILE T cell activation, increased late expression -64- L25444 TAF2E TATA box binding protein (TBP)-associated factor M95787 TAGLN transgelin NM_005421 TAL2 T-cell acute lymphocytic leukemia 2 L47345 TCEB3 transcription elongation factor B (11OkD, elongin A) M57732 TCF1 hepatic nuclear factor (HNFl) NM_003205 TCF12 helix-loop-helix transcription factors 4 M96956 TDGF1 teratocarcinoma-derived growth factor 1 U19878 TMEFF1 transmembrane with EGF and follistatin like M92357 TNFAIP2 tumor necrosis factor, alpha-induced protein 2 M59465 TNFAIP3 tumor necrosis factor, alpha-induced protein 3 X83490 TNFRSF6 tumor necrosis factor receptor member 6 U37518 TNFSFIO tumor necrosis factor member 10 NM 003294 TPSB1 tryptase beta 1 U19261 TRAF1 TNF receptor-associated factor 1 U78798 TRAF6 TNF receptor-associated factor 6 S69790 WASF3 WAS protein family, member 3 U53476 WNT7A wingless-type MMTV integration site family L15309 ZNF141 zinc finger protein 141 (clone pHZ-44) U78722 ZNF165 zinc finger protein 165 HG4333- ZNF79 zinc finger protein 79 (pT7) HT4603 X57809 lambda light chain variable region HG3111- Homo sapiens clone HH409 unknown HT3287 U79249 Human clone 23839 sequence AB000464 clone:RES4-24A HG4593- voltage-gated sodium channel (SCN1A) HT4998 X77744 Homo sapiens for FLJO0032 protein, partial U79248 Human clone 23826 sequence AI420129 ESTs Method of identifying genes regulated by the IL-18 are disclosed in Example 3. These studies showed that IL-18 is a bona fide proinflammnatory cytokine and can directly regulate the expression of several genes encoding other proinflammatory mediators. Studies using human 5 blood samples show that many responses to IL-18 occur widely in the human population and demonstrate their utility as biochemical markers of IL-18, and consequently anti-IL1 8, function. Modulators of IL-18 can be agonists and antagonist. Preferably the modulator is a binding protein or a neutralizing binding protein. Exemplary IL-18 inhibitors include, but are not limited to, antibodies, and fragments 10 thereof, that bind to IL-18; antibodies that bind to IL-18R; antibodies that bind to IL-18RAcP; IL 18bp; IL-18R fragments (e.g., a solubilized extracellular domain of the IL-18 receptor); peptides that bind to IL-18 and reduce or prevent its interaction with IL-18R; peptides that bind to IL-18R and reduce or prevent its interaction with IL- 18 or with IL-1 8RAcP; peptides that bind to IL -65- 18RAcP and reduce or prevent its interaction with IL-18R; and small molecules that reduce or prevent IL-18 production or the interaction between any of IL-18, IL-18R, and IL-18RAcP. Certain IL-18 inhibitors are described, e.g., in US Pat. No. 5,912,324, issued July 14, 1994; EP 0 962 531, published Dec. 8, 1999; EP 712 931, published Nov. 15, 1994; US Pat. No. 5 5,914,253, issued July 14,1994; WO 97/24441, published July 10, 1997; US Pat. No. 6,060,283, issued May 9, 2000; EP 850 952, published Dec. 26, 1996; EP 864 585, published Sep. 16, 1998; WO 98/41232, published Sep. 24, 1998; US Pat. No. 6,054,487, issued April 25, 2000; WO 99/09063, published Aug 14,1997; WO 99/22760, published Nov. 3, 1997; WO 99/37772, published Jan. 23,1998; WO 99/37773, published March 20, 1998; EP 0 974 600, published Jan. 10 26, 2000; WO 00112555, published Mar.9, 2000; Japanese patent application JP 111,399194, published Oct. 31, 1997; Israel patent application IL 121554 AO, published Feb. 8, 1998; which are incorporated herein by 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 made using 15 suitable equivalents without departing from the scope of the invention or the embodiments disclosed herein. Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting of the invention. 20 Examples Example 1: Production and Characterization of recombinant IL-18 Example 1.1: Assays to determine biological activity of IL-18 Throughout Example 1 and Example 2 the following assays were used to determine biological 25 activity of IL-18 unless otherwise stated. Example 1.1.A: KG-1 Bioassay KG-1 (ATCC #CCL-246) is a human myelomonocytic cell line that constitutively expresses low levels of functional IL-18 receptor. Treatment with TNF up-regulates both the IL 18Rc. and 0 subunits of the functional IL-18 receptor on these cells. The KG-i bioassay was 30 performed by incubating TNF-treated KG-I cells with recombinant human IL-18 (rhu-IL-18) and determining the level of IL-18-induced human IFNy production by an ELISA. (Konishi, K., et al (1997) J. Immunol. Methods 209:187-191). The KG-I bioassay was used to determine the neutralization potency of IL-18 antagonists. For example, anti-IL-18 antibodies were incubated -66with different concentrations of rhu-IL-18 and then incubated with TNF-treated KG-I cells in a 96-well plate for 16-18 hours at 37*C. The supernatants collected and assayed for human IFNy levels by an ELISA. This assay can measure IC50 values down to 4x10- 11 - 6x10-II M of an IL 18 antagonist. 5 Example I.1.B: Human Whole Blood Assay Briefly, the Human Whole Blood Assay (WBA) determines neutralization potency of IL 18 antagonists against natural IL-18 within a physiological context. In this assay, the readout was inhibition of endogenous IL-i 8-dependent human IFNy production. Whole blood was stimulated with LPS (1 gg/mL) plus IL-12 (50 pg/mL) in the presence or absence of IL-18 antagonists at 10 37*C. Human IFNy concentrations were determined by ELISA 18-24 hrs post-LPS plus IL-12 stimulation. Example 1.1.C: Receptor Binding Assay Briefly, in the Receptor Binding Assay (RBA), '2I labeled rhu-IL-18 was used to determine binding of IL-18 to IL-18 receptor. '2I-rhu-IL-18 binds specifically to the IL-18RaDf 15 on TNF-treated KG-i cells (-7,000 sites/cell). 12I-rhu-LL-18 has the same specific activity as unlabeled IL-18 and can be competed off by unlabeled IL-18. Two modes of inhibition, A and B, were defined. In neutralization Mode A , binding of IL-18 to the high affinity IL-18 receptor (IL-18Rca0) was not effected, but IL-I8-mediated signal transduction (i.e. IFNy production) was blocked. In neutralization Mode B binding of IL-18 to 20 IL-18Rc4D was blocked and thereby no subsequent receptor-mediated signaling occured. Example 1.2: Production of recombinant IL-18 Example 1.2.A: Plasmid construction, Expression and purification of human prolL-18 25 Recombinant human IL-1 8 was generated by expressing the precursor form of IL-18 in SF-9 insect cells. Using standard molecular biological methods well known in the art, full-length human pro-IL-18 cDNA was generated using specific PCR primers based on published sequence (Ushio, S., et al. (1996) J. Immunol. 156:4274-4279) and subsequently cloned into the baculovirus (BV) transfer vector pVL1393. (BD Biosciences, San Jose, CA; Cat# 51-21201P) 30 The 5' PCR primer used to generate full-length human pro-IL-18 cDNA contained sequences -67encoding a 6-Histidine region such that the N-terminus of the proIL-18 contained a 6-HIS-tag. SF9 insect cells were infected with baculovirus harboring pVL1393 vector containing IL-18 cDNA. Infected SF9 cells were lysed and the lysates were run over a nickel column to purify recombinant HIS-tagged proIL-18 (rhu pro IL-18). (BD Biosciences, San Jose, CA; Cat# 554802 5 ) The recombinant HIS-tagged prolL-18 was processed further by digesting with human caspase I to generate biologically active IL-18 (mature IL-18). (Ghayur T., (1997) Nature 386:619-623). Example 1.2.B: NEM treatment of IL-18 Recombinant human IL-18 obtained from Hayashibara Biochemical Laboratories, Japan, 10 displayed batch variation in specific activity and in IL-18 binding affinity. IL-18 contained disulphide bonds between various pairs of the four cysteines in mature IL-18. These caused structural and functional heterogeneity, and variations between batches. Homology modeling of human IL-18 using IL-lb coordinates showed that cysteine residues at positions 38 and 68 of mature human IL-18 are exposed and therefore reactive. 15 Recombinant human IL-I8 from Example 1.2.A was treated with N-ethyl maleiamide (NEM) to protect the cysteines from oxidation. NEM-IL-18 was monomeric, did not form aggregates, was stable, and retained high specific activity over time. NEM-IL-18 retained neutralizing epitopes because anti-huIL-18 neutralizing antibodies bound and neutralized NEM IL-18. Despite NEM treatment of IL-18, neutralizing epitopes on NEM-IL-18 were preserved as 20 determined by the ability of anti-IL-18 antibodies to neutralize biological activity of both NEM IL-18 and natural human IL-18 in human WBA. NEM-IL-18 was used for assay optimization and selection and initial characterization of fully human anti-human -IL-18 mAbs. Example 1.2.C: Generation and characterization of 4C/A mutant of IL-18 The 4C/A mutant of IL-18 was generated by mutating the four Cysteine residues in 25 mature IL-18 to Alanine ("4C/A-huL-18"). Comparison of 4C/A mutant of IL-18 with NEM huIL-18, as summarized in Table 4 below, showed that the two proteins were indistinguishable in biological and biochemical properties. Both 4C/A mutant of IL-18 and NEM-huIL-18 were monomeric by dynamic light scattering (DLS) and size exclusion chromatography (SEC) analysis, and similar in conformation and physical stability by circular dichroism analysis. The 30 biological activity of 4C/A mutant of IL-I8 and NEM-huIL-1 8 were the same in the KG-i assay, and both forms of IL-18 bound IL-18BP and anti-IL-18 antibodies with similar affinity. 4C/A huL-18 was not subject to oxidative instability and was readily expressed at high levels in E. coli. -68- Table 4 Comparison of NEM-IL-18 and 4C/A-huIL-18 shows that they are equivalent in measurements of conformational and oligomeric purity, physical stability, and binding to antibodies or cell-bound receptors. Properties Measurements NEM-huIL-18 (4C/A)-huIL-18 Oligomeric state SEC Monomeric Monomeric DLS Monomeric Monomeric Confirmation CD (wavelength CD minimum at 210 nm CD minimum at 210 nm scan) Stability CD (temperature Stable up to 400 C Stable up to 40* C scan) Bioactivity IFNy production by 8 ng/mL IFNy 8 ng/mL IFNy 2 ng/mL IL-18 Epitopes Neutralization of Neutralized by IL-18BP- Neutralized by IL-18BP IFNy production by Fc, 125-2H and IL-18Ra Fc, 125-2H and IL-I8Rx reference binder Biacore (KD) IL-18BP-Fc: 0.098 nM IL-18BP-Fc: 0.135 nM 125-2H: 0.2 nM 125-2H: 0.2 nM 2.5(E)mgl: 0.3 nM 2.5(E)mgl: 0.2 nM 5 Example 1.2.D: Generation and characterization of Biotinylated rhuIL-18 (biot-IL-18) Biotinylation of NEM-IL- 18 from Example 1.2.B was performed on lysine residues using standard techniques well known in the art, (Sulfo-NHS-LC-Biotin, Pierce, Rockford, IL; Cat#21335), and the biotinylated rhu-IL-18 (biot-IL-18) obtained was a heterogeneous mixture 10 containing species with 1, 2, 3, or 4 biotins per huIL-18. Furthermore, species with 2 or 3 biotins per rhuIL-18 were the major species in biot-IL-18. Biot-IL-18 was biologically active, bound anti IL-18 antibodies as determined by ELISA, and was neutralized by all neutralizing anti-huIL-18 antibodies tested. Biot-IL-18 bound KG-1 cells expressing IL-18Ra4 on their surface with high affinity, and the biot-IL-18 on the surface of KG-1 cells was detected by FACS analysis using -69anti-biotin antibodies (Sigma-Aldrich, St Louis, MO; cat# B 3640 ). Thus biotinylation does not interfere with receptor binding and does not mask neutralizing epitopes of rhuIL-18. Example 1.2.E: Generation and characterization of 1251 Labeled rhuIL-18 The lysine residues on NEM-JL-18 from Examplel.2.B were labeled with 12I using 5 conditions specified by Amersham (Piscataway, NJ; Cat # IM5861 ). "2 I-labeled IL-18 retained its specific activity, was competed by non-modified IL-18, and bound specifically to IL-18R on KG-i cells. Binding of ' I-labeled IL-18 to IL-18 receptor was blocked by neutralizing anti huIL-18 monoclonal antibodies. Thus iodination did not affect receptor binding of IL-18 and did not mask neutralizing epitopes on the IL-18. 12I-labeled IL-18 was used to determine the 10 neutralization mode and potency of anti-IL-18 antibodies in the Receptor Binding Assay. EXAMPLE 2: Generation and isolation of anti IL-18 antibodies Example 2.1: Assays to identify anti-IL-18 antibodies 15 Throughout Example 3 the following assays were used to identify and characterize anti IL-18 antibodies unless otherwise stated. Example 2.1.A: ELISA An ELISA was developed to screen for antibodies that bind human IL-18. In this ELISA, biotinylated NEM-huIL-18 (see Example 1.2.B) was captured by either goat anti-biotinylated IgG 20 or on to streptavidin coated plates. Hybridoma or B cell supernatants were applied and IL-18 bound antibodies were detected using HRP-conjugated anti-human IgGs, following standard ELISA protocols well known in the art. Example 2.1.B: Affinity Determinations using BIACORE technology The BIACORE assay (Biacore, Inc, Piscataway, NJ) determines the affinity of antibodies 25 with kinetic measurements of on-, off-rate constants. Antibodies are captured on a biosensor chip by means of a covalently linked secondary antibody (e.g. goat anti-human IgG or anti-mouse IgG) and then varying concentrations of recombinant IL-18 are applied. Binding is recorded as a function of time and kinetic rate constants are calculated. In this assay, on-rates as fast as 10 6
M
's-' and off-rates as slow as 10~6 s-4 can be measured. 30 -70- Example 2.1.C: Epitope Mapping BIACORE technology was used for mapping the epitopes recognized by IL-18 antagonists such as anti-IL-18 antibodies. Briefly, one IL-18 antagonist was captured on the Biacore chip and rhuIL-18 was bound to the immobilized reagent. Binding of another anti-IL-18 5 antagonist to this complex was then tested. Simultaneous binding of two reagents demonstrates that the two recognize distinct epitopes. Example 2.2: Generation of Anti-IL-18 HuMAbs Using XENOMOUSE The XENOMOUSE transgenic mouse technology (Abgenix, Inc., Fremont, CA) was 10 employed to obtain fully human anti-human IL-18 monoclonal antibodies (HuMAbs). This technology consists of transgenic mice carrying human variable heavy chain locus carrying VH, DH, and JH, Cmu, Cdelta and a single human lgG constant heavy chain locus and light chain gene loci. Upon immunization with an antigen of interest, these mice generate fully human antibodies to the antigen. 15 Example 2.2.A: Immunization of XENOMOUSE with IL-18 antigen XENOMOUSE animals were immunized via footpad route for all injections. Total volume of each injection was 50ul per mouse, 25ul per footpad. Initial immunization injection contained 40ug human IL-18 (NEM-rhuIL-18) in pyrogen-free DPBS admixed 1:1 v/v with TiterMax Gold per mouse. Subsequent boosts were made with 40ug Human IL-18 in pyrogen 20 free DPBS admixed with 25ug of Adju-Phos (aluminum phosphate gel) per mouse for six times, then a final boost of 40ug Human IL-18 in pyrogen free DPBS without adjuvant per mouse. The animals were immunized on days 0, 4, 8, 11, 17, 21, 25 and 35 for this protocol. Fusions were performed on day 39. Following the immunization regimen described above, mice were euthanized, then inguinal and Lumbar lymph nodes were recovered. 25 Example 2.2.B: Generation of Hybridoma Lymphocytes were released by mechanical disruption of the inguinal and Lumbar lymph nodes, obtained according to Example 2.2.A, using a tissue grinder, and depleted of T cells by CD90 negative selection. Hybridoma fusion was performed by mixing washed enriched B cells and non-secretory myeloma P3X63Ag8.653 cells purchased from ATCC, cat. # CRL 1580 30 (Kearney et al, J. Immunol. 123, 1979, 1548-1550) at a ratio of 1:1. The cell mixture was gently pelleted by centrifugation at 800 g. After complete removal of the supernatant, the cells were -71treated with 2-4 mL of Pronase solution (CalBiochem, San Diego, CA; cat. # 53702; 0.5 mg/m-l in PBS) for no more than 2 minutes. Then 3-5 ml of FBS was added to stop the enzyme activity and the suspension was adjusted to 40 ml total volume using electro cell fusion solution, ECFS (0.3M Sucrose, Sigma-Aldrich, St Louis, MO; Cat# S7903, 0.1mM Magnesium Acetate, Sigma, Cat# 5 M2545, 0.1mM Calcium Acetate, Sigma-Aldrich, St Louis, MO; Cat# C4705). The supernatant was removed after centrifugation and the cells were resuspended in 40 ml ECFS. This wash step was repeated and the cells again were resuspended in ECFS to a concentration of 2x10 6 cells/mi. Electro-cell fusion was performed using a fusion generator, model ECM2001, Genetronic, Inc., San Diego, CA. The fusion chamber size used was 2.0 ml using the following instrument settings: 10 Alignment condition: voltage: 50 v, time: 50 s; Membrane breaking at: voltage: 3000 v, time: 30 rs; Post-fusion holding time: 3 s. After fusion, the cells were resuspended in hybridoma fusion medium: DMEM (JRH Biosciences), 15 %FBS (Hyclone), containing 0.5XHA (Sigma-Aldrich, St Louis, MO; cat. # A9666), and supplemented with L-glutamine, pen/strep, OPI (oxaloacetate, pyruvate, bovine 15 insulin) (all from Sigma) and IL-6 (Boehringer Mannheim, Indianapolis, IN) for culture at 37 *C and 10% CO 2 in air. Cells were plated in flat bottomed 96-well tissue culture plates at 4x10 4 cells per well. Cultures were maintained in hybridoma fusion medium for 2 weeks before transfer to Hybridoma medium: DMEM (JRH Biosciences, Lenexa, KS), 15 %FBS (Hyclone, Logan, Utah), and supplemented with L-glutamine, pen/strep, OPI (oxaloacetate, pyruvate, bovine 20 insulin) (all from Sigma) and IL-6 (Boehringer Mannheim, Indianapolis, IN). Hybridomas were selected for by survival in 0.5XHA hybridoma fusion medium and supernatants from those wells containing hybridomas were screened for antigen reactivity by ELISA. The ELISA format entailed incubating supernatants on antigen coated plates (human IL-18 coated plates) and detecting human anti-human IL-18 binding antibodies using horseradish peroxidase (HRP) 25 labeled mouse anti-human IgG, then the all positive samples were confirmed by two sets of ELISA in parallel, which entailed incubating supernatants on antigen coated plates (human IL-18 coated plates) and detecting human anti-Human IL-18 binding antibodies using horseradish peroxidase (HRP) labeled mouse anti-human Gamma and Kappa chain. Cloning was performed on selected antigen-positive wells using limited dilution plating. 30 Plates were visually inspected for the presence of single colony growth and supernatants from single colony wells then screened by antigen-specific ELISAs as described above. Highly reactive clones were assayed to verify purity of human gamma and kappa chain by multiplex ELISA using a Luminex instrument. -72- Example 2.2.C: XENOMAX Technology Alternatively, lymphocytes obtained in Example 2.2.B were subjected to Selective Lymphocyte Antibody-generation Method (SLAM), which defines XENOMAX antibody selection technology (Abgenix, Inc., Fremont, CA). Single B cells were plated in 96 well plates B 5 cells producing human monoclonal antibodies to desired antigen (human IL-18) were identified by a plaque forming cell assay (Babcook, J.S., Leslie, K.B., Olsen, O.A., Salmon, R.A., and Schrader, J.W. Isolation of functional antibody genes from single lymphocytes of defined antigen-specificity. Proc. Natl. Acad. Sci. USA, 93:7843-7848, 1996) and IgG genes were cloned by single-cell RT-PCR of isolated B cells using 5' primers for VH and Vk leader 10 sequences and 3' primers specific for human Cgamma and Ckappa. The recombinant IgG genes obtained were expressed in mammalian cells as described in Examples 2.2.E and 2.2.G. Example 2.2.D: Identification of anti-IL-18 antibodies Hybridomas and B-cell producing antibodies that bound IL-18, generated according to Examples 2.2.B and 2.2.C, were identified using biotinylated IL-18 ELISA (see Example 2.1.A). 15 Hybridomas and B-cell supernatants containing antibodies that bound IL-18 were then tested for IL-18 neutralization potency in the KG-1 bioassay performed according to Example 1.1.A. Neutralizing anti-IL-18 antibodies (from hybridoma and SLAM aproaches) were subcloned into a mammalian expression vector, expressed in COS cells, purified and re-tested in the KG-i bioassay (seeTable 5). 20 Table 5 Neutralization Potency of Anti-IL-18 HuMAbs in KG-1 Bioassay HuMAb# KG-1 Assays (IC 50 ,M) NEM-rhulL-18 Hybridoma 2.5.1 3E-10;4E-10 2.13.1 2E-10; IE-10; 7E-11 2.3.3 1E-9; 2E-10; 7E-10 XENOMAX -73- 215 lE-10; 3E-10; 1E-10; 444 lE-10; 2E-10; 2E-10 478 7E-10; 2E-9; 3E-10 435 8E-10; 7E-10; 4E-10; 413 IE-9; 7E-10; 7E-10 581 7E-10; 3E-10; 3E-9 231 IE-10; 3E-11; 2E-9 521 6E-10; 3E-10; 2E-9 336 7E-10 351 2E-10 490 5E-10 550 TBD 268 7E-9 The variable regions of the antibodies in Table 5 are described inTablel Example 2.2.E: Subloning of neutralizing anti-IL-18 HuMAbs into mammalian 5 expression vector Genes for the heavy and light chains of antibodies were cloned in pCDNA (Invitrogen, Carlsbad, Ca) vectors under control of the CMV promoter following manufacturer's instruction. These plasmids with human genomic Gamma-2 and kappa sequences were used to cotransfect COS cells by electroporation with heavy and light chains corresponding to each clone employing 10 standard conditions well known in the art. Cells were allowed to recover, grow, and secrete antibodies for 72 hours in serum free DMEM supplemented with glutamine. Culture supernatants were then collected, clarified by centrifugation and filtration, and put over Protein-A resin. Columns were washed with PBS, antibodies eluted with low pH buffer, and quickly neutralized with IM Tris solution. Antibody 15 preps were buffer-exchanged with PBS on Amicon-30 spin filters. Concentration and purity of antibodies were analyzed by spectrometry at OD 280 and SDS-PAGE before they were tested for IL-18 neutralization potency. To achieve greater expression levels of human antibodies in COS cells, the heavy and light chains of some antibodies were subcloned into vector pEF-BOS (Mizushima, S. and Nagata, 20 S. (1990) Nucleic acids Research Vol 18, No. 17) under control of elongation factor promoter. In short, PCR primers for heavy chain variable regions were designed in such a way that they could be inserted into a cassette pEF-BOS plasmid containing an IgG signal peptide and the -74sequence of human IgGI constant region [wild type (SEQ ID No. 2) or inactive mutant (SEQ ID No. 3)]. The forward VH PCR primer contained restriction site Nrul, as did the nucleotide sequence of the signal peptide. The reverse VH PCR primer contained SalI restriction site that was also engineered into the 5' end of the gamma-I Fc sequence. The Vi 1 PCR fragments were 5 digested with NruI/SalI and cloned into the pEF-BOS human IgGI wild type or pEF-BOS human IgGimutant constructs. The entire light chain genes were moved into pEF-BOS vector in their existing Kappa format from pCDNA vectors by HindIII restriction digest, filling the overhangs with T4 polymerase, followed by NotI digest. These blunt/NotI light chain fragments were then cloned into SrfINotI digested pEF-BOS vector. 10 VH and VL regions of antibodies were cloned from original hybridoma lines. RNA was prepared from antibody-producing cells, RT-PCR performed with primers designed as described above, i.e NruI/SalI primers for VH, and Nrul/BsiWI primers for VL. The full length IgG1 and Kappa chains were assembled into cassette vectors. The selected antibodies were further modified. Naturally occuring antibodies have either 15 glutamine (Q) or glutamate (E) as the heavy and/or light chain NH2-terminus. Production of antibodies with Q as the NH2-terminus yields NH2-terminal heterogenity due to the cyclization of the glutamine residue to a glutamate. Therefore, the glutamine residue at the NH2-terminus of some of the antibodies was mutated to glutamate. Also two residues, Leucine234 and Leucine235 in the hinge region of the Fc portion, were mutated to prevent the effector functions of the 20 antibody. Briefly, Leucine 234 and Leucine 235 were each replaced by an Alanine residue using standard molecular biological techniques( Lund, J. et al., J. Immunology (1991) 147: 2657-2662; Winter, el al. US PAT NOS 5,648,260; 562482 1;5,624,821) . These Fc-mutated antibodies were termed (mgl). These mutants are further characterized in Examples 2.2.J 6, below. 25 Example 2.2.F: Characterization of Selected Neutralizing anti-IL-18 antibodies Several recombinant anti-human IL-18 antibodies with distinct germ line sequences were produced in mammalian cells, purified and functionally characterized in various assays (see Table 6). 30 -75- Table 6: In vitro antigen binding, cell assay and backbone sequence characteristics of some anti-IL-18 antibodies. Biacoreb RBA KG-1b WBA Gene Familiesc Antibody Technology" (KD, nM) (IC 5 0 , nM) (IC 50 , nM) (IC 50 , nM) Sequence Diversity 2.5(E)mgl Hybridoma 0.31 2.38 0.20 3 VL-L2 VH5-51 2.5(E)wtgl Hybridoma 0.40 2.38 0.30 3 VL-L2 VH5-51 215(E)mgld Xenomax 0.23 1.17 0.17 3 VL-A27 VH4-31 444(Q)mgld Xenomax 1.61 2.49 0.13 1 VL- VH4 A27(7) 31(1) 581(E)mgld Xenomax 2.00 1.28 1.3 3 VL-A2 VH3-30 2.3.1(E)wtgl Hybridoma 0.23 0.20 0.63 2 VL-02 VH4-59 2.13.1(E)wtgl Hybridoma 0.20 0.20 0.12 2 VL- VH4 A27(8) 31(18) NEM-cys protected rhuIL-18 was used. 5 Numbers in parenthesis indicate differences in amino acids from clone closet to germ line sequence Example 2.2.G: Generation of CHO Cell Line Producing 2.5(E)mgl Stable CHO cell lines expressing 2.5 (E)mgl antibody were generated following the procedures outlined below. 10 Example 2.2.G 1: Construction of Expression Vector The plasmid pA510 was constructed for high-level expression of antibodies in mammalian cell lines. This pUC19-derived plasmid contained the E. coli ColEi origin of replication and the beta-lactamase gene for ampicillin resistance. Briefly, cDNA corresponding to the VH and VL regions of the 2.5(E)mgl antibody were 15 cloned using standard molecular biological techniques, fused to mutated human gamma-1 and kappa constant region genes, respectively, such that DNA encoding a native, fully-human, IgG1/kappa antibody was produced. These DNAs were introduced into expression construct pA5 10. The resulting plasmid contained sequences (exclusive of pUC19) for the following genes or regulatory elements in the following order: 5'-CMV enhancer, adenovirus major late promoter, 20 human immunoglobulin signal peptide, 2.5(E)mgl heavy-chain immunoglobulin variable region, -76human gamma-i immunoglobulin constant region, SV40 polyadenylation sequence, human gastrin transcription termination sequence, SV40 origin of replication (SV40 promoter/enhancer), murine dihydrofolate reductase sequence, thymidine kinase polyadenylation sequence from Herpes Simplex virus, CMV enhancer, adenovirus major late promoter, human immunoglobulin 5 signal peptide, 2.5(E)mgl light-chain immunoglobulin variable region, human immunoglobulin kappa constant region, and SV40 polyadenylation sequence -3'. The coding regions were inserted downstream from strong viral promoters that drove the antibody gene transcription. The vector also encoded the expression of the mouse DHFR gene, which enabled selection of transformed cells by virtue their ability to grow in culture in the absence of nucleosides. 10 Example 2.2.G 2: Transfection of Expression Vector Into Parental Cell Line The cell line, CHO DUX B 11. (Urlaub, 0. and Chasin L.A. Proc Natl Acad Sci USA 77: 4216-4220 (1980)), defective in the expression of the dihydrofolate reductase (DHFR) gene, was used for transfection of the expression vector described in Example 2.2.G 1. CHO DUX B 11 15 cells were transfected with the vector using calcium phosphate precipitation method well known in the art (Current Protocols in Molecular Biology; Ausubel, F.V., Brent, R., Moore, D.M., Kingston, R.E., Seidman, J.G., Smith, J.A., 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 plate. The plates were incubated at 37 0 C for two hours. One hundred and fifty 20 micrograms of DNA were dissolved in 2.7ml water in a 50ml conical tube. Three hundred microliters of 2.5 M CaC12 was added and this DNA mix 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 vortexed for 5 seconds and incubated at room temperature for 20 minutes. One ml was distributed evenly over each plate (still in F12) and the plates were 25 incubated at 37 0 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 continued for one minute after which the DMSO was diluted by the addition of 5 ml of phosphate buffered saline (PBS) to each plate. The plates were aspirated and washed two more times in PBS. Ten ml of Gibco alpha MEM with nucleosides was added and the plates were incubated at 37 0 C overnight. The next day, the 30 medium was changed to Gibco alpha MEM without nucleosides with 5 % dialyzed fetal bovine serum (FBS), and six hours later the cells were seeded into 96-well plates as follows. The cells from the 10cm 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 seeded at 10 mI/plate, 100 I/well. One hundred ml of the same medium was added to the remaining 100 -77ml of cells and 20 additional 96-well plates were seeded as above. (This was a second dilution.) The medium was changed in the 96-well plates one week later and again a week after that. The alpha MEM medium without nucleosides was used to select cells expressing DHFR and therefore the expression vector. 5 Example 2.2.G 3: Selection of 2.5(E)mgl producing cells Culture supernatants from transfected CHO cells were tested for the presence of secreted antibody 2.5(E)mgl using an ELISA specific for human IgG. Once a set of CHO transfectants had been screened for expression of human antibody, an additional selection was used to isolate 10 those cells that had amplified the number of copies of the expression vector integrated in the CHO genome. The drug methotrexate (MTX) was used for the selection of amplified lines. Cultures grown in the presence of MTX were tested for their ability to produce inmnunoglobulin. The MTX-resistant lines that expressed more antibody than their MTX-sensitive predecessors were taken through another cycle of selection in higher concentration MTX, and tested for 15 immunoglobulin production. 2.5(E)mgl expressing CHO cells were cultured in a 1 or 15 liter bioreactor and the yield of antibody was determined to be - 1.0g/L in a two-week run. Example 2.2.H: Physicochemical Characterization of CHO Cell-Derived 2.5(E)mg1 Preliminary physical and chemical characterization of CHO derived 2.5(E)mgl was 20 performed. The experimentally determined molecular weight of 2.5(E)mgl 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)mgl had the correct N termini for both light and heavy chains. There was very little heavy chain C terminal variability, 25 as 99% of the 2.5(E)mgl molecules lacked lysine at the heavy chain carboxy termini. Each 2.5(E)mgl heavy chain contained a single N-linked glycosylation site with oligomannose and complex, fucosylated binatennary structures with 0, 1 or 2 terminal galactose residues. Example 2.2.1: Solubility and Stability of CHO Cell-Derived 2.5(E)mg1 30 Purified 2.5(E)mgl was soluble to at least 62 mg/mL in pH 5, 6 and 7 buffers for a minimum of 4 weeks. Accelerated stability studies with 2.5(E)mgl at 37*C in these buffers were performed to identify stability-indicating assays and the optimal long-term storage pH. Samples were taken at weekly intervals for analysis by size exclusion HPLC and SDS-PAGE to test for aggregation and fragmentation, LC-MS/MS peptide mapping for S-S bond detection, antigen -78- ELISA and/or cell based bioassay for activity measurement, and cation exchange HPLC and iso Asp quantification for charge heterogeneity measurement. Preliminary analysis of the samples by SEC (size exclusion chromatography), SDS-PAGE and cation-exchange chromatography showed that all three assays indicated stability and therefore 2.5(E)mgl is more stable at -pH6. 5 Example 2.2.J: Characterization of CHO-cell-derived anti-IL-18 HuMAb, 2.5(E)mgl Example 2.2.J 1: IL-18 Species Specificity The ability of 2.5(E)mgl to bind and/or neutralize IL-18 from human, cynomolgus monkey, mouse, rat and dog was evaluated. Using the BIACORE assay following manufacturers 10 intructions (see Example 2.1.B), it was shown that 2.5(E)mgl bound mature human IL-18, but not mouse IL-18. In addition, immunoprecipitation data showed that 2.5(E)mgl bound cynomolgus monkey IL-18 (IC50 for cyno 1L-18= 9.1E X 101), but not dog or rat IL-18. 2.5(E)mgl functionally neutralized human and cynomolgus IL-18 bioactivity in a similar manner, but no inhibition of dog, rat or mouse IL-18 was seen. 15 Example 2.2.J 2: Human Cytokine Specificity The specificity of 2.5(E)mgl for IL-18 was evaluated using the BIACORE assay following manufacturers intructions (see Example 2.1.B). The 2.5(E)mgl antibody was captured on the biosensor chip and its ability to bind a panel of known human cytokines in solution was determined. As shown in Table 7, 2.5(E)mgl bound recombinant human mature IL-18 and pro 20 IL-18. In contrast, 2.5(E)mgl did not bind to any of the other 23 human cytokines tested, including the IL-I family members IL-la and IL-I .. Table 7 Biacore Analysis of Cytokine Binding by 2.5(E)mgl Captured 2.5(E)mgl (25 mg/mL) Soluble rec. human cytokines, (1gM) 2.5(E)mgl Binding IFNy IL-Ia IL-1 Other cytokines' IL-18b+ Pro-IL-18 + -79- " Additional cytokines tested for binding included IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-21, TNF, LT, LTalD2, and LTa2$1. 2.5(E)mgl did not bind to any of these cytokines. Cysteine > Alanine mutant BV derived rec. human IL-18 5 Example 2.2.J 3: Affinity Measurements Table 8 shows the in vitro IL-18 binding properties of 2.5(E)mgl measured using the BIACORe assay according to manufacturer's instruction. The 2.5(E)mgl antibody had a fast on rate, slow off-rate with an overall affinity of 0. 196nM. The kinetic rate parameters of two 10 reference IL-18 antagonists (125-2H and IL-18 binding protein) are shown for comparison. Table 8 IL-18 Binding Properties of 2.5(E)mgl and Reference Reagents Reagent Biacore Parameters On-rate Off-rate KD (x 101 M-Is- 1 ) (x 10-6 s- 1 ) (nM) 2.5(E)mg1" 268 52.4 0.196 Murine anti-human IL-18 (125-2H)b 190 110 0.550 IL-18BP-Fcc 140 26 0.190 (4C/A)-HuIL-18 was tested in BIACORE. b -125-2H, a neutralizing mouse anti-human IL-18 IgGI mAb ' -IL-18BP-Fc, an Fc fusion of the natural IL-18 antagonist 15 Example 2.2J 4: In Vitro IL-18 Neutralization Potency The in vitro neutralization potency of 2.5(E)mgl was determined in the KG-i bioassay, the receptor binding assay (RBA) and the human WBA (see Example s 1.1.A-1.1.C). As shown in Table 8 the 2.5(E)mgl antibody neutralized both recombinant (KG-1 and RBA) and natural IL-18 20 (WBA), (IC 50 <0.5 nM in KG-1, <2 nM in RBA and <5 nM in WBA), and is consistent with its IL-18 binding affinity. -80- Table 8 Neutralization Potencies of 2.5(E)mgl and Reference Reagents Neutralization Potency in vitro (IC50, nM) Reagent KG-1" RBAb WBAC 2.5(E)mgl 0.2 2.4 3.0 Murine anti-human IL-18 0.2 >300' 3.0 mAb (125-21H) IL-18BP-Fc 0.03 1.0 5.7 Anti-IL-18R mAb (M-840) 1.5 1.7 2.7 b KG-1 bioassay, mean values (4C/A)-huLL-18 was used in this assay d Receptor Binding Assay 5 Human Whole blood assay with 4-6 individual donors. Mean values given. 125-2H neutralizes IL-1 8 bioactivity despite failure to inhibit receptor binding Example 2.2.J 5: In Vivo Neutralization Potency of 2.5(E)mg1 To evaluate the ability of 2.5(E)mgl to neutralize natural human IL-18-induced IFNy in 10 an inflammatory environment in vivo, the severe combined immunodeficient (SCID) mouse model was used, wherein human PBMCs were injected into the mouse and the cells were stimulated in vivo to produce human IL-18 (HuPBMC-SCID model). The results (Table 9) showed that 2.5(E)mgl inhibited human IL-18-dependent human IFNy production in vivo with a clear dose-response by either route of administration. The ED 5 0 of 2.5(E)mgl was approximately 15 1 pg or 0.1 jig / per mouse (=0.05 mg/kg or 0.005 mg/kg) by ip or iv administration, respectively. 20 -81- Table 9A In vivo efficacy of 2.5(E)mgl administered i.p. in HuPBMC-SCID mouse model Group hulFNg (pgIml) % Inhibition 2.5(E)mgl 0.025 j.g/mouse 70 +17 61 2.5(E)mgl 0.25 jg/mouse 112 +29 36 2.5(E)mgl 2.5 jig/mouse 36 +10 80 2.5(E)mgl 25 pg/mouse 10 8 94 2.5(E)mgl 250 jig/mouse 3 +2 98 No Treatment 193 +59 HuIgG Control 250 jig/mouse 177 433 Table 9B In vivo efficacy of 2.5(E)mgl administered i.v. in HuPBMC-SCID mouse model Group huIFNg (pgl) % Inhibition 2.5(E)mgl 0.025 jig/mouse 156 +45 36 2.5(E)mgl 0.25 jg/mouse 27 +9 89 2.5(E)mgl 2.5 jig/mouse 36+ 8 85 2.5(E)mgl 25 jig/mouse 11 +6 96 2.5(E)mgl 250 jg/mouse 4 +2 98 No Treatment 279 + 26 HuIgG Control 250 jig/mouse 245 +22 5 Example 2.2.J 6: Effector Functions The Fc portion of an antibody mediates several important effector functions e.g. cytokine induction, antibody-dependent cell-mediated cytotoxicity (ADCC), phagocytosis, complement dependent cytotoxicity (CDC), and half-life/clearance rate of antibody and antigen-antibody 10 complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives. Certain human IgG isotypes, particularly IgG1 and IgG3, mediate ADCC and CDC via binding to FcyRs and complement Clq, respectively. Neonatal Fc receptors (FcRn) are the critical components determining the circulating half-life of antibodies -82- The L234A and L235A mutations in 2.5(E)mgl did not influence the overall affinity or the neutralization potency of 2.5(E)mgl HuMAb as compared with 2.5(E)wtgl (Table 10). However, as expected, these mutations did abolish binding to FcyR and Clq. 5 Table 10: Mutations of residues L234 and L235 to Alanine does not affect affinity or neutralization potency of 2.5(E)mgl Kinetic Rate Parameters KG-1 Bioassay Ab On-rate Off-rate KD IC 5 0 (103 M's-1) (x 10-6 s-') (nM) (nM) 2.5(E)wtgl 281 47.8 0.170 0.4 2.5(E)mgl 268 52.4 0.196 0.2 Example 2.2.J 6.1: FcyR I binding The human FcyR I (CD64) has a relatively high affinity for IgGI immune complexes (KD 10 1E-8-IE-9 M). It is expressed on monocytes and macrophages and a number of myeloid cell lines including U937. The binding of 2.5(E)wtgl and 2.5(E)mgl to U937 cells was determined by fluorescence-activated cell sorting (FACS) (CURRENT PROTOCOLS IN IMMUNOLOGY. Vol (1) 5.3.1, Edited by J. E. Coligan et. al., Published by John Wiley & Sons, Inc., 2002). The data obtained (see Table 11) demonstrated that 2.5(E)wtgl binds to U937 cells, but as expected, 15 2.5(E)mgl did not. To confirm that this binding was mediated through FcyR I, a mouse anti hFcyR I blocking antibody (10.1) was used for competition experiments. The result showed that antibody 10.1 blocked binding of 2.5(E)wtgl to U937 cells in a dose-dependent manner at the concentrations tested below, and thus, 2.5(E)wtgl binds FcyR I on U937 cells. 20 Table 11. Demonstration of the failure of 2.5(E)mgl, in contrast to 2.5(E)wtgl, to bind FcyR I on U937 cells (data shown as MFI+/-SD) Antibody 1.OOE-09 1.00E-08 1.00E-07 1.OOE-06 1.00E-05 1.0OE-04 1.OOE-03 1.00E-02 1.00E-01 Conc (AM) 2.5(E)wtgl 0.50+0.00 0.50+0.00 0.63+0.12 0.57+0.05 0.60+0.00 1.10+0.00 5.80+0.05 29.60+0.05 38.76+5.19 2.5(E)mgl 0.67+0.09 0.67+0.09 0.60+0.00 0.80+0.14 0.63+0.05 0.67+0.05 0.53+0.05 0.60+0.05 0.63+0.08 -83- Example 2.2.J 6.2:FcyR II binding The human FcyR II (CD32) has a relatively low binding affinity for IgGI immune complexes (KD 1E-5-1E-7M). Under physiological conditions, it requires the formation of 5 multivalent immune complexes for activation. Using fluorescein isothiocyanate (FITC) labeled antibodies specific for FcyR I, II or III and detection by flow cytometry, we validated the expression of FcyR II on K562 cells and used this cell line for the FcyR II binding assay. The binding of monomeric 2.5(E)wtgl to K562 cells was very weak. Therefore, an anti-kappa chain antibody was used to pre-crosslink the IgG1 molecules to mimic multivalent immune complexes 10 and tested their binding to FcyR II on K562 cells. After cross-linking, 2.5(E)wtgl bound to K562 cells, but even after cross-linking 2.5(E)mgl showed only minimal, if any, binding (Table 12). An anti-FcyR II antibody, clone IV3, blocked binding of 2.5(E)wtgl, and thus, the 2.5(E)wtgl binding to K562 was FcyR II mediated. Table 12. Binding of 2.5(E)wtgl and 2.5(E)mgl to FcyR II on K562 cells 15 after crosslinking (data shown as MFI+/-SD) Antibody 1.OOE-08 1.OOE-07 1.OE06 1.OOE-05 1.OOE-04 1.O0E-03 1.OOE-02 1.OOE-01 I.OE+00 Conc (PNI) 2.5(E)wtgl 0.37+0.05 0.37+0.05 0.40+0.00 0.43+0.05 0.80+0.08 3.43+0.21 19.7+0.70 93.33+4.90 134.37+12.93 2.5(E)mgl 0.30+0.00 0.40+0.00 0.40+0.00 0.37+0.05 0.37+0.05 0.40+0.00 0.50+0.00 1.60+0.08 5.37+0.38 Example 2.2.J 6.3:Clq binding Complement activation and lysis of the cells via the classic pathway is activated through 20 binding of Clq to the Fc portion of IgG molecule. The binding of Clq to 2.5(E)wtgl and 2.5(E)mgl was determined using standard ELISA techniques known in the art (Hezareh, M., et. al., (2001) J. Virology,75 (24):12161-12168). 2.5(E)wtgl and 2.5(E)mgl HuMAbs were coated onto plastic plates followed by incubation with human Clq. Bound Clq molecules were then detected by a mixture of goat-anti-human Clq and rabbit anti-goat IgG alkaline phosphate 25 conjugate. The results showed that 2.5(E)wtgl bound Clq, but the 2.5(E)mgl did not (Table 13). -84- Table 13. Demonstration of the failure of 2.5(E)mgl, in contrast to 2.5(E)wtgl, to bind to C1q by ELISA (data shown as OD4 0 s+/-SD) C1q Conc 0 20 40 60 80 100 120 (pg/m) 2.5(E)wtgl 0.09+0.00 0.78+0.00 0.98+0.00 1.06+0.07 1.14+0.06 1.32+0.13 1.24+0.06 2.5(E)mg1 0.10+0.01 0.12+0.00 0.16+0.00 0.18+0.00 0.21+0.01 0.21+0.01 0.22+0.00 5 Example 2.2.J 6.4: Neonatal Fc Receptor (FcRn) Binding Interaction of IgG with the neonatal Fc receptor (also called Brarible receptor) in endothelial cells has been proposed to be an IgG quality control system and the critical determinant for the long half-life of IgGs [ Ghetie, V., et al (1997) Nat. Biotechnol. 15:637-640). IgG molecules taken up by pinocytosis and binding successfully to FcRn in endocytic vacuoles 10 are returned to circulation. IgG molecules that fail to bind to FcRn are degraded. The critical residues of human IgG for FcRn binding have been mapped to the junction of the CH2-CH3 domains (Kim J.K., et al (1999):Eur. J. Immunol. 29:2819-2825 ). Importantly, these FcRn binding residues are conserved between human and mouse immunoglobulins and human immunoglobulins bind to mouse FcRn allowing structure activity relationship studies in 15 mice. To test the effect of the L234A and L235A mutations on FcRn binding, binding of the wild-type 2.5(E)wtgl and the mutant 2.5(E)mgl to FcRn in vitro was determined using a FcRn expressing CHO cell line. 2.5(E)wtgl and 2.5(E)mgl were incubated with the FcRn expressing CHO cells at pH 6.5, followed by incubation with FITC-conjugated anti-human IgG (20 Ab). The 20 cells were washed and analyzed by FACS. The 500 nM concentration of 2.5(E)mgl and 2.5(E)wtgl showed significant binding to the FcRn compared to the 0.5 nM concentration, which was similar to background with cells alone. 25 Example 2.2.K: Pharmacokinetics in Mice Pharmacokinetics (PK) of 2.5(E)mgl were assessed in a screening mouse study to determine if the Fc mutations (L234A, L235A) introduced to prevent binding of 2.5(E)mgl to FcyR and Clq adversely affected the serum PK profile. The mouse FcRn bound mouse and human IgG equally -85well making the mouse a relevant species for structure activity relationship studies in mice (Ober, R.J., et al (2001) Int. Immunol. 13:1551-1559). In mice, the terminal half-life of 2.5(E)mgl was estimated to be 12 days. In similar studies, the half-lives of other human monoclonal antibodies were 10 - 14 days. 5 The pharmacokinetics of 2.5(E)mgl were evaluated in female mice (Jackson Labs, C57BL/6n) following a single intravenous dose of 0.2 mg (equivalent to an average of 10 mg/kg). A total of 24 mice were dosed and 3 samples were drawn from each mouse. The sampling scheme extended through seven days. 2.5(E)mgl exhibited a distribution phase followed by an elimination phase. The distribution and elimination half-life estimates were approximately 1.6 10 hours and 12 days, based on a two compartment open model (Table 14). Table 14 Summary of key pharmacokinetic parameters of 2.5(E)mgl derived from a single intravenous dose in mice t1/2a t1/2p Cmax CL Vs 5 VI V 2 MRT AUC (hr) (days) (g/mL) (mL/hr) (mL) (mL) (mL) (days) (br* Lg/mL) 1.58 12.2 63.2 0.0162 6.82 3.15 3.67 17.5 12250 15 Disease models Example 2.2.L: Effect of anti-IL-18 antibodies in disease models Example 2.2.L.1: Inhibition of LPS-induced IFNg production by anti-mulL-18 mAbs LPS-induced IFNy production is dependent upon IL-18 expression (Ghayur, T., et al, 20 1997. Nature 386:619-623. ). An LPS-induced IFNy production assay was used to determine the efficacy of 93-10C to inhibit IL-18-dependent LPS-induced IFNy production in vivo. Mice were given a single iv dose of 93-10C (50 jig). Thirty minutes later mice were challenged with LPS (20 mg/kg) and bled 4h later. Serum IFNy titers were determined by ELISA. As shown in Table 15, 93-1OC inhibited LPS-induced IFNy production by -70%. 25 Table 15 93-10C inhibits LPS-induced IFNg production in vivo Group muIFNg (pg/nil) % Inhibition Rat IgG 250 jg/mouse 7239+ 365 N/A MBT 93-1OC 250 pg/mouse 2395 +711 67 -86- Example 2.2.L.2: Inhibition of carrageenan-induced paw edema IL-18 is involved in neutrophil recruitment to sites of inflammation. Carrageenan induced foodpad edema is a monocyte and neutrophil-dependent inflammation model. Edema in 5 this model can be significantly inhibited by neutralizing the biological activity of IL-18 (Leung, B.P., et al (2001) J. Immunol. 167:2879-2886 ). Mice were dosed (ip) with 1C5 (400 gg) (Hyashibara Laboratories, Japan) or 93-10C (100 gg) (Medical and Biological Laboratories (MBL) Co. Watertown MA.)or control antibodies and then injected with carrageenan (sc) in the hind footpads. Carrageenan-induced edema was measured daily from 24 h to 96 h. IC5 and 93 10 10C significantly suppressed carrageenan-induced edema (-50% inhibition) from 24h to 96 h post challenge (Table 16). In addition to blocking neutrophil infiltration, 93-1OC also blocks TNF expression at the site of inflammation in this model (Leung, B.P., et al (2001) J. Immunol. 167:2879-2886). ). Table 16 In vivo suppression of carrageenan-induced paw edema 15 Carrageenan Chan e in Paw Swelling (nun) Time (hrs) 24 48 72 96 125-2H @ 400 0.357 0.557 0.543 0.414 1C5 @ 400 ug 0.214 0.300 0.286 - 0.200 Rat IgG @ 100 ug 0.300 0.500 0.550 I 0.450 93-10C @ 100 ug 0.157 0.271 0.243 0.157 P<0.05 Vs. Control IgG Example 2.2.L.3: Collagen-induced arthritis Rheumatoid arthritis (RA) is characterized by chronic inflammation of joints, and, loss of bone and articular cartilage. Although RA is thought to be an autoimmune disease, the 20 autoantigen involved has not been identified and the precise etiology of disease is unknown. Collagen-induced arthritis (CIA) is a widely used model of RA and has histopathological features which are similar to the human disease (Bendele, A., et al (1999) Toxicol Pathol. 27:134-142; Trentham, D.E. et al (1977) J. Exp. Med. 146:857-868). In this model, genetically-susceptible mice or rats are immunized with type II collagen (CII) in complete Freund's adjuvant. The 25 resulting polyarthritis is characterized by destruction of cartilage, bone resorption, synovitis, and periarticular inflammation (Bendele, A., et al (1999) Toxicol Pathol. 27:134-142 ). IL-18 KO -87mice on a DBA/1 background showed decreased incidence and severity of CIA when compared to wild-type mice (Wei, X.Q., et al (2000) J. Irnmunol. 166:517-521 ). To address the role of endogenous IL-18 in the pathogenesis of CIA, mice were treated with a rabbit polyclonal IgG (BA77) that neutralizes mouse IL-18. When dosed for 14 days from 5 the time of priming, BA77 delayed disease onset and resulted in a significant decrease in disease severity. BA77 also significantly inhibited production of IgG2a anti-collagen antibodies. These results are similar to those reported for IL- 18 KO mice and confirm a role for IL-18 as an important proinflammatory cytokine in early CIA. The data from IL-18 KO mice and anti-IL-18 IgG treated wild type mice indicate that 10 IL-18 plays an important proinflammatory role during CI-induced primary T cell activation. To better understand the role of IL-18 during the onset of CIA, mice were immunized with CI and treatment with rat IgG or 93-10C initiated just prior to disease onset, which occurs around day 14. Treatment with 93-IOC resulted in a significant delay in disease onset and severity when compared to control rat IgG (Table 17). These data show that IL-18 is a significant factor not 15 only in T cell priming but also in promoting arthritogenic responses after activation of CII specific T cells. Table 17 Anti-IL-18 mAb 93-10C delayed the onset and decreased the severity of CIA Treatment Mean Arthritic Score Day 11 12 13 14 '15 f .:1 18.
Rat IgG @200 jig 0.00 0.13 0.13 0.13 0.27 0.53 1.20 1.20 93-10C @ 200 jig 0.00 0.00 0.00 0.00 0.07 0.00 0.20 0.47 Dexamethasone-21-P @lmpk 0.00 0.00 0.27 0.27 0.13 0.13 0.13 0.13 Treatment Period P<0.05 vs. Rat lgG 20 Treatment Mean Arthritic Score Day 19 20 21 22 23 25 26 27 Rat IgG @200 [tg 1.53 1.73 1.93 2.27 2.53 4.20 4.27 4.53 93-IOC @ 200 gg 0.47 0.80 1.00 1.00 1.13 2.20 2.27 2.87 Dexamethasone-21-P @lmpk 0.07 0.07 0.07 0.00 0.00 0.00 0.07 0.20 Treatment Period P<0.05 vs. Rat IgG -88- Example 2.2.L.4: Septic Arthritis IL-18 is an important factor in the pathogenesis of a mouse model of septic arthritis. This is generally not thought to be a model of RA, but shares some inflammatory components and 5 pathology related to RA. In this model, disease is induced by injecting live group B streptococci (GBS) into knee joints. The severity of ensuing arthritis correlates with both systemic and local levels of IL-1P and IL-6, but not TNF (Tissi L., et al (1999) Infect. Immunol. 67:4545-50). Significant IL-18 levels in the joints were detected as early as 12 hours post injection with serotype IV (GBS) followed by peak IL-18 production after 5 days (-550 pg/ml in IC5 treated vs 10 -30 pg/mi in IgG control). Elevated IL-18 levels were detected in the serum by day 5 post injection (-180 pg/nl in IC5 treated vs -20 pg/ml in IgG control). When IC5 was injected 1 hour prior to administration of GBS there was marked inhibition in the frequency of articular lesions from day 2 through day 10 (arthritic index: 1.0 in IC5 treated vs 2.5 in IgG control). In addition, IC5 treatment also resulted in significant 15 reduction in cytokine levels in the joints including IL-6 and IL-1p. (Data not shown) Example 2.2.L. 5: SLE The most studied models of lupus involve strains of mice (MRL/lpr and NZB/NZW F1) 20 that spontaneously develop lupus-like syndrome with severe glomerulonephritis, autoantibody production (anti-DNA, anti RNP etc.), splenomegaly, lymphadenopathy, and to some extent arthritis and vasculitis. Kidney involvement is observed usually at 3-5 months of age, progresses rapidly, and by 6-10 months is fatal. Both mouse strains have been extensively studied to gain an understanding of clinical disease. 25 The NZB/NZW Fl (B/W) mouse model (The Jackson Laboratory, Maine, USA) was selected as the most relevant model to evaluate the effects of exogenous IL-18 on lupus-like disease progression. The onset of disease progression in B/W mice is observed usually at 7-9 months of age and by 12-14 months is fatal as a result of renal failure. To investigate the role of IL-18 in lupus pathogenesis, B/W mice were treated daily with r-mulL-18 or vehicle control 30 beginning at 7 months of age. Kidney function was assessed by determining the degree of proteinuria. Daily treatment of B/W mice with 50pg/kg of IL-18 led to accelerated onset of severe proteinuria as compared to the PBS vehicle treated group. IL- 8 treated B/W mice also -89exhibited accelerated deaths. These observations were consistent with those described above for the MRIJlpr mice and underscore a pro-inflammatory role for IL-18 in autoimmune disease. To investigate the therapeutic effect of IL- 18 blockade in a mouse model of SLE, an induction-maintenance treatment protocol in B/W mice that recapitulates clinical therapy for 5 lupus nephritis was established. In this study, severely nephritic B/W mice received 5 weekly doses of cytoxan (induction phase) followed by chronic IC5 or mouse IgGI control (125-2H) treatment (maintenance phase). Results demonstrate that in the ensuing 130 days of maintenance treatment, IC5 significantly prolonged survival of BW mice as compared to control IgGI 125-2H. 125-2H is a 10 mouse IgGI mAb that does not recognize inull.-18 (P< 0.05,). In addition to the prolongation of survival, the onset of severe proteinuria was delayed, and there was a reduction of IgG2a and IgGI anti-dsDNA in 1C5 treated BW mice. The reduction of anti-ds DNA by 1C5 treatment was transient and not statistically significant. Antibody against IC5 (mouse anti-mouse antibody [MAMA]) was detected, and preceded the loss of efficacy after day 130, as evidenced by a 15 precipitous drop in survival and the loss of effect in the reduction of anti-ds DNA titer and proteinuria. In conclusion, despite the presence of antibody responses to 1C5, IL-18 blockade by IC5 prolonged survival, delayed onset of severe proteinuria, and reduced anti-ds DNA titers in B/W mice. These data demonstrate a role for IL-18 in promoting inflammatory responses resulting in a loss of kidney function and ultimately death. 20 Example 2.2.L. 6: Multiple Sclerosis The contribution of IL-18 to the pathogenesis of experimental allergic encephalomyelitis (EAB; a murine model of MS) was investigated. Relapsing-remitting EAE is considered to be a relevant model for the human disease due to similar disease course, clinical signs, and CNS 25 pathology. In these studies the disease was induced in IL-18 KO mice and WT C57/B16 mice. The IL-18 KO mice showed a slight delay in onset of disease symptoms compared to WT mice, and developed significantly less severe disease at later time points (Table 18). Treatment of WT mice with BA77 (anti-mouse IL-18 IgG (250 mg, 2X / wk), at day 0 through day 14 post immunization delayed the onset of disease symptoms and significantly limited disease severity at 30 later time points (Table 18). The protective effect of anti-IL-18 IgG could be observed at later time point even after cessation of treatment. -90- Table 18 Anti-IL-18 Ab treated mice and IL-18 knockout mice develop less severe EAE disease Mean Clinical Score Day 14 Post Immunization PLP Induced EAE IgG(BA77) 4 in SJL/J mice anti-IL-18 ab 2.7 Mean Clinical Score Day 18 MOG Induced EAE WT 3.6 in IL-18 KO and WT mice KO 2.1 5 Example 2.2.L.7: Liver Damage Concanavalin A (Con A)-induced liver inflammation/damage is an animal model of T-cell-mediated liver disease. Activation of intra-hepatic T-cells by Con A leads to local production of inflammatory mediators (e.g. IFNy and Fas ligand). Fas-Fas ligand interaction results in production of IL-18 that induces further IFNy, Fas ligand and TNF production. Thus, a 10 positive feedback loop is established that results in liver damage and excessive production of liver enzymes such as ALT and AST from dying cells. IC5 or 93-10C mAbs were injected (ip) lh prior to iv administration of 150 pg of Con A. Mice were bled 24h post Con A injection and serum titers of liver enzymes (ALT & AST) were determined. Both IC5 and 93-IOC blocked LPS-induced elevation of liver enzymes, although 93-10C was effective at lower doses (Table 15 19). Table 19 In vivo inhibition of ConA-induced liver inflammation by 93-10C Treatment AST stdv ALT stdv PBS 57 16 30 2 ConA alone 1138 416 1294 481 ConA+93-10C (50ug) 183 70 153 88 ConA+93-10C (12.5ug) 635 427 443 256 ConA+rat IgGl(50ug) 3924 __ 1062 3455 753 -91- Example 2.2.L.8: Sensis IL-18 has emerged as an important mediator of endotoxic shock. IL-18 may be a critical mediator of endotoxin-induced lung, liver and multi-organ failure (Neeta, M.G., et al (2000) J. Immunol. 164:2644-2649). This effect of IL-18 may be dependent upon its ability to regulate 5 production of cytotoxic mediators as well as its ability to activate innate immune responses and recruit neutrophils to the site of local inflammation. In addition, LPS challenge induces elevated serum levels of IFNy, TNF and IL-1 and these cytokines may contribute towards LPS-induced lethality. IL-18 knockout mice challenged with LPS were deficient in LPS-induced IFNy and produce significantly less TNF and IL-I than WT mice (Takeda, K., et al. (1998) Immunity 10 8:383-390). LPS induced lethality experiments were performed as follows. Animals were weighed on Day 0 and the appropriate dosage to be administered was determined. At T = -1 hour, animals were injected with anti-IL-18 antibodies or control antibodies in 500sl of 0.9% saline, Intra peritoneal (IP). At T = 0, animals were injected with 20 mg/kg lipo poly saccharide (LPS) ( E. 15 coli serotype 0111:B4 Sigma Cat #L-4130 lot #71K4110) in 100tl of 0.9% saline, Intra-venous (IV). Four hours later blood was obtained from the animals via cardiac puncture. Serum mulFNy titer was determined by muIFNy ELISA (R&D Systems). WT mice treated with anti-muIL-18 mAbs, iC5 or 93-10C, were protected from LPS induced lethality (Table 20) (125-2H, which has the same inactive isotype as IC5 but does not 20 bind mulL-18, served as the control). In addition, anti-IL-18 IgG treated mice were reported to have reduced lung and liver damage after LPS challenge and this correlated with reduced neutrophil accumulation(Neeta, M.G., et al (2000) J. Immunol. 164:2644-2649). Table 20 1C5 and 93-10C mAbs prevent high dose LPS lethality 25 Lethality Percent Survival Time (hrs) 0 8 24 32 48 56 72 120 144 Saline 100 100 100 50 10 10 10 10 10 125-2H @ 400 100 100 80 70 10 10 10 10 10 ICS @ 400 pg 100 100 100 90 80 80 80 80 80 Lethality Percent Survival Time (hrs) 0 8 24 32 48 56 72 120 144 Saline 100 100 90 40 10 10 10 10 10 Rat IgG @ 100 g 100 100 100 100 70 40 40 40 40 93-10C @ 100 g 100 100 100 100 100 100 100 100 100 -92- Example 2.2.M: Crystallization of 2.5(E) Fab fragment To demonstrate that the antibodies of the invention can be crystallized such that formulations and compositions comprising crystallized antibody can then be generated the following experiments were undertaken. 5 Example 2.2.M.1: Preparation and Purification of the 2.5(E) Antibody Fab Fragment The 2.5(E) human IgG was expressed in CHO cells in SR-286 Media. The supernatant after lysis was filtered through a 0.5 micron filter and loaded onto a Protein A column pre equilibrated in Protein A Buffer A (lXPBS). The IgG was then eluted with Protein A Buffer B (0.1 M Na Acetate pH 3.5, 150 mM NaCl). The pooled IgG was concentrated to 20 mg/ml, mixed 10 with 50% papain gel slurry, and incubated at 37*C for 24 hours with vigorous shaking. The antibody/slurry mixture was then dialyzed against 50 mM Tris buffer pH 7.0 overnight at 4'C to remove cysteine from the buffer. A 25 mL Protein A Sepharose 4 Fast Flow affinity column (Amersham Pharmacia) was prepared by washing with with 100 mL of Buffer A (50 mM Tris pH 7.0). The dialyzed supernatant was applied to the affinity column (2 mLJmin flow rate). 15 2.5(E)Fab fractions (monitored by UV absorbance at 280 nm) were collected in the flow-thru. Fractions containing a 2.5(E)Fab concentration greater than 0.3 mg/mL (determined by UV absorbance at 280 nm) were pooled and concentrated to -20 mg/mL using an Ultrafree-15 Biomax 10 kDa molecular weight cut-off (MWCO) centrifugal filter device (Millipore) and frozen at -80 *C. This concentrated sample was used in crystallographic experiments described 20 below. Sample purity was assessed with SDS-PAGE. Example 2.2.M.2: Crystallization of the 2.5(E)Fab Fragment Frozen 2.5(E)Fab stock solution (-20 mg/mL) was thawed on ice. The Fab (2 pL) was mixed with 2 gL of a reservoir solution consisting of 25-30% polyethyleneglycol (PEG) 400, 100 25 mM CAPS pH 10.5 and suspended over the reservoir on the underside of a siliconized glass cover slip at about 4 *C. Rod-like crystals appeared within one day. The rod-like crystals were determined to be 2.5(E) Fab fragment crystals (Data not shown). Example 3: IL-18 responsive genes 30 Example 3.1: Materials and Methods Throughout Example 4, the following materials and methods are used unless otherwise stated. -93- Example 3.1.A: Cell Treatment and RNA Preparation Example 3.1.A.1: KG-1 cells Approximately 3.0 x 10 7 KG1 cells (ATCC #CCL-246) were used for each experimental 5 condition in four treatment groups. In the first, cells were treated with 50 ng/mL recombinant IL18 with or without a 30 min. preincubation with 10 mg/mL cyclohexinide. After 30 min. or two hours cells were harvested for RNA. In the second, cells were treated with 0, 0.5, 2.0, 10 or 50 ng/mL recombinant IL18 with or without a 30 min. preincubation with 10 mg/mL cycloheximide. After two hours cells were harvested for RNA. In the third, cells were treated 10 with 0 or 10 ng/mL TNF. Following an overnight incubation, cells were then treated with 0, 0.5, 2.0, 10 or 50 ng/mL recombinant IL18 with or without a 30 min. preincubation with 10 mg/mL cycloheximide. After two hours cells were harvested for RNA. In the final treatment group, cells were treated simultaneously with 0 or 10 ng/mL TNF and 0 or 2.0 ng/mL recombinant IL18 with or without a 30 min. preincubation with 10 mg/mL cycloheximide. After two hours cells were 15 harvested for RNA. Total RNA was prepared using TRIZOL Reagent (Life Technologies, Rockville, MD). An initial phase separation was performed according to the manufacturer's protocol and was followed by an additional extraction using a half volume of phenol: chloroform: isoamyl alcohol (25:24:1, Life Technologies, Rockville, MD). RNA precipitation and wash were performed 20 according to the manufacturer's TRIZOL protocol instructions. Approximately 3 micrograms of RNA were electrophoresed on a 1.0% agarose/formaldehyde denaturing gel to assess quality. For experiments requiring TNF preincubation, KG-i cells were incubated for 12 hours with 2 ng/mI TNF prior to stimulation with 2, 10 or 40 ng/ml of IL18. RNA was prepared as described above. 25 Example 3.1.A.2: Human whole blood assays 2.5mL human whole blood was aliquoted into 15mL conical tubes and treated with IL18, L12, IL18+IL12, IL18+IL12+anti-IL18 or IL18+IL12, IL18+IL12+control antibody. Final concentrations were as follows: IL12-500pg/mL, IL18(YK27-1)-50ng/mL, migG-5ug/mL, anti 30 IL18 1252H-5ug/mL, and anti-IL18 2.5-4ug/mL. Mixtures were incubated at 37'C for four hours with gentle intermittent inversion. After incubation, red blood cells were removed using ammonium chloride by adding 5mL lXlysis buffer (PharM Lyse Ammonium Chloride Lysing Reagent diluted 1:10 in Depc). After 5 minutes on ice the mixture was centrifuged at 1200 rpm for five minutes. This procedure was repeated once yielding a white pellet of blood leukocytes. -94- RNA was isolated subsequently using the Trizol procedure described above. For micro array analysis, all sample volumes were increased by a factor of four. Example 3.1.B: Preparation of Probe and Target Hybridization 5 Ten micrograms of total RNA and the SuperScript Choice System for cDNA Synthesis (Gibco BRL, Gaithersburg, MD) were used to synthesize double stranded cDNA. The synthesis was carried out according to Affymetrix (Santa Clara, CA) protocol, which requires T7-(dT) 4 oligomer primers (GENSET) in place of the oligo (dT) or random primers provided with the kit and incubations at 42*C during the temperature adjustment and first strand synthesis steps. The 10 resulting cDNA was cleaned with Phase Lock Gel Light 2 ml tubes (Eppendorf AG, Hamburg, DE), and the pellet was suspended in 12 pL of DEPC-H 2 0. 5 pL of the cDNA was used in conjunction with the BioArray High Yield RNA Transcript Labeling Kit (Enzo, Farmingdale, NY) to produce biotin-labeled cRNA targets by in vitro transcription (IVT) from T7 RNA polymerase promoters. Free nucleotides were removed from the IVT reaction with RNeasy Mini 15 Columns (Qiagen, Hilden, DE). 15 jig of biotin-labeled cRNA was then fragmented according to Affymetrix protocol. The entire fragmented sample was combined with 5 pL control oligonucleotide B2 (Affymetrix), 15 gL 20X Eukaryotic Hybridization Control (Affymetrix), 3 pL sonicated salmon sperm DNA (10 mg/mL, Stratagene, La Jolla, CA), 3 pJL acetylated BSA (50 mg/mL, Gibco BRL), 150 p.L 2X MES hybridization buffer, and water to a final volume of 20 300 p.L. Following Affymetrix protocol, Genechip HuGeneFL Arrays (Affymetrix) were pre-wet with IX MES. The hybridization cocktails were then heated and centrifuged, and 200 pL was loaded onto the chips. The chips were spun in a 45*C rotisserie oven for 16 hours. Example 3.1.C: Washing, Staining, and Scanning Probe Arrays 25 The hybridization cocktail was removed from the chips and replaced with a non-stringent wash buffer. Chips were washed and stained using the EukGE-WS2 protocol on the GeneChip Fluidics Station 400, according to manufacturer's instruction (Affymetrix); a protocol that stained the chips with both Streptavidin Phycoerythrin (SAPE) stain solution and antibody solution. All necessary wash buffers and stains were prepared according to Affymetrix protocols. 30 A GeneArray Scanner (Agilent, Palo Alto, CA) was used in conjunction with GeneChip software (Affymetrix) to scan the stained arrays. -95- Example 3.1.D: Data Analysis Genechip data was transferred from Affymetrix MAS4 to Microsoft Excel then uploaded into Spotfire Decisionsite 7.0. 5 Example 3.2: Gene expression regulated by IL-18 Example 3.2.1: IL18 alone directly regulates a cohort of genes in KG1 cells. To determine transcripts regulated directly by IL18, cytokine titration experiments were performed using KG1 cells in the presence and absence of the protein synthesis inhibitor cycloheximide. Shown in Table 1 is a list of 62 transcripts represented by 67 different probe sets 10 (due to redundancies on the chip) found to have be regulated two fold or more with a p value of less than 0.05 (using Student'st test) under at least one condition in the presence and absence of cycloheximide. These genes comprised a variety of functional categories including transcription factors, kinases, and secreted proteins. Because these genes are regulated without de novo protein synthesis, these genes respond directly to ILl 8 induced signaling. Twelve genes encode secreted 15 proteins, and thirteen encode surface molecules (making these feasible antibody targets). The remaining genes encode nuclear and cytoplasmic proteins (see Table 21). -96- Table 21. Genes induced by IL18. Genbank ID Location Gene Name Unigene Comment 0.5 2 10 50 /function _ _ _ ng/m l n/m ng/m ng/mil L29217 kinase CLK3 CDC-like kinase 3 9.1 7.4 8.1 15.0 D14497 kinase MAP3K8 mitogen-activated protein kinase kinase kinase 8 6.6 2.9 5.8 3.9 L19871 neither ATE3 activating transcription factor 3 1.0 1.1 3.3 2.6 U15460 neither BATF basic leucine zipper transcription factor, ATF-like 1.5 1.7 2.4 2.8 U45878 neither BIRC3 baculoviral 1AP repeat-containing 3 7.0 6.2 10.2 10.0 U37546 neither BIRC3 baculoviral LAP repeat-containing 3 29.4 26.9 76.6 63.6 U72649 neither BT32 BTG family, member 2 3.1 4.7 6.6 5.9 107765 neither CESI carboxylesterase 1 1.0 1.3 2.1 2.1 M27691 neither CREBI cAMP responsive element binding protein 1 0.9 2.4 4.9 3.1 HG3548- neither CUTL1 cut (CCAAT displacement protein) 2.5 2.1 1.3 0.7 HT3749 X59131 neither D13SI06E highly charged protein 2.1 0.5 1.5 2.3 U53445 neither DOCI downregulated in ovarian cancer 1 2.0 3.3 3.0 3.8 X68277 neither DUSPI dual specificity phosphatase 1 2.5 3.1 4.1 3.3 U48807 neither DUSP4 dual specificity phosphatase 4 2.0 2.3 2.9 2.0 X52541 neither EGRI early growth response 1 15.5 12.7 32.4 20.3 X63741 neither BGR3 early growth response 3 5.9 7.3 15.1 9.0 L07077 neither EHADH enoyl-Coenzyme A 3.4 2.3 1.8 2.5 M62831 neither ETR101 immediate early protein 3.4 5.8 6.3 6.8 LI 9314 neither HRY hairy (Drosophila)-homolog 2.3 2.5 2.3 2.0 581914 neither IBR3 immediate early response 3 17.0 18.6 32.9 29.6 X51345 neither JUNB jun B proto-oncogene 7.2 6.1 10.7 9.6 U20734 neither JUNB jun B proto-oncogene 10.2 21.8 25.0 25.4 U49957 neither LPP LIM domain-containing 2.2 1.1 2.0 1.9 M58603 neither NFKBI nuclear factor kappa B (p105) 1.6 2.0 2.9 2.3 S76638 neither NFKB2 nuclear factor kappa B 1.7 2.2 3.5 4.3 M69043 neither NFKBIA nuclear factor kappa B 9.6 10.4 15.5 15.8 U91616 neither NFKBIE nuclear factor kappa B 11.6 14.8 20.7 21.0 L13740 neither NR4AI nuclear receptor subfamily 4, group A. member 1 2.0 2.7 2.4 2.5 HG4115- neither ORIE3P olfactory receptor 4.5 12.0 4.2 4.1 HT4385 L20971 neither PDB4B phosphodiesterase 4B, cAMP-specific 2.4 2.8 4.2 3.5 U64675 neither RANBP2LI RAN binding protein 2-like 1 1.1 1.8 2.2 2.2 S57153 neither RBBP1 retinoblastoma-binding protein 1 2.5 3.4 5.0 4.1 X75042 neither REL v-rel 1.6 2.5 3.9 3.7 M83221 neither RELB v-rel 2.3 2.8 2.8 2.6 S59049 neither RGSI regulator of C-protein signalling 1 10.9 12.7 22.4 17.8 U70426 neither ROS16 regulator of CG-protein signalling 16 3.9 4.7 7.5 6.7 U22377 neither RLF rearranged L-myc fusion sequence 2.5 2.0 2.5 2.6 M95787 neither TAGLN transgelin 6.6 4.7 1.0 1.6 L47345 neither TCEB3 transcription elongation factor B (1 OkD, elongin 3.6 5.3 2.3 4.2 A) M59465 neither TNFAIP3 tumor necrosis factor, alpha-induced protein 3 9.9 12.4 25.4 20.6 U19261 neither TRAF TNFreceptor-associated factor 1 2.8 2.8 4.9 4.1 U78798 neither TRAF6 TNF receptor-associated factor 6 1.2 2.0 2.1 2.2 M37435 secreted CSFI colony stimulating factor I (macrophage) 2.9 2.9 2.1 2.6 M57731 secreted GRO2 GRO2 oncogene 15.2 20.9 26.3 27.0 X53800 secreted GRO3 GRO3 oncogene 4.1 5.5 14.8 9.9 X04500 secreted IL1B interleukin 1, beta 2.2 3.4 5.7 4.7 M28130 secreted IL8 interleukin 8 6.2 10.0 13.4 14.5 Y00787 secreted 118 interleukin 8 5.8 7.4 8.3 8.5 U89922 secreted LTB lymphotoxin beti (TNF superfamily, member 3) 5.0 5.7 11.0 12.8 -97- Genbank ID Location Gene Name Unigene Comment 0.5 2 10 50 /function ng/nl nWnl ng/i ng/ml M31166 secreted TX3 pentaxin-related gene, rapidly induced by IL-I 3.1 5.2 10.3 6.4 beta M23178 secreted SCYA3 small inducible cytokine A3 1.8 2.0 5.0 3.8 M69203 secreted SCYA4 small inducible cytokine A4 0.9 1.9 7.0 5.6 J04130 secreted SCYA4 small inducible cytokine A4 1.0 2.6 5.9 4.5 M92357 secreted TNFAIP2 tumor necrosis factor, alpha-induced protein 2 4.2 6.4 20.3 19.3 Z32765 surface CD36 CD36 antigen (collagen type / TSP receptor) 1.6 2.0 1.4 1.2 ZI 1697 surface CD83 CD83 antigen 4.7 8.2 19.6 16.7 M57730 surface BFNA1 ephrin-Al 9.8 6.0 9.5 15.2 A28102 surface GABRA3 gamma-aminobutyric acid (GABA) receptor 3.0 2.5 1.6 2.7 M24283 surface ICAMI intercellular adhesion molecule 1 (CD54) 7.5 11.5 14.5 13.9 M55024 surface ICAMI intercellular adhesion molecule 1 (CD54) 2.5 3.4 3.2 3.7 J03171 surface IFNARl interferon (alpha, beta and omega) receptor 1 3.2 2.5 2.8 2.6 X01057 surface IL2RA interleukin 2 receptor, alpha 0.7 0.4 3.9 3.6 L10338 surface SCN1B sodium channel polypeptide 1.8 2.3 1.5 1.5 D79206 surface SDC4 syndecan 4 (amphiglycan, ryudocan) 4.0 4.2 7.2 6.1 HG961- surface SoSi son of sevenless (Drosophila) homolog 1 6.3 6.2 9.1 9.9 HT961 X83490 surface TNFRSF6 tumor necrosis factor receptor member 6 1.1 1.3 3.8 3.3 U19523 neither ICHI GTP cyclohydrolase 1 2.1 U37518 surface ITNFSFIO tumor necrosis factor member 10 1.4 1.4 2.3 1.6 Example 3.2.2: Cytokine exposure history effects KG-1 cell response to IL-18. Since cytokines typically appear sequentially during an immune response, the effect of 5 preincubating KG-1 cells with TNF prior to treating with ILl 8 were tested. This experiment also tested the hypothesis that the cytokine exposure history of cells may effect their response to subsequent cytokine exposure. Cells were treated with 2ng TNF 12 hours prior to adding IL18 and harvested four hours later. IL1 8 regulated the expression of approximately 125 genes under these conditions (Table 10 2). The filtering criteria used to obtain this set of genes was less than 50% change due to TNF and a two fold or greater change due to IL8 at the 10 ng/mL and 40 ng/mL. These genes comprised a variety of functional categories including transcription factors, kinases, and secreted proteins (Table 22). In contrast to other conditions tested here, we find interferon gamma mRNA and protein to be induced by IL18 following exposure to TNF. 15 -98- Table 22. Genes regulated by IL18 following TNF treatment. Genbank ID Gene Name Unigene Comment Fold 10 ng Fold 40 ng J00219 IFNG interferon, gamma 26.3 31.8 U17034 PLA2RI phospholipase A2 receptor 1, 180kD 29.6 28.7 M57710 LGALS3 lectin, galactoside-binding, soluble, 3 (galectin 3) 27.5 25.4 X97748 PTX3 pentaxin-related gene, IL-e induced 15.2 13.6 M27288 OSM oncostatin M 23.1 12.0 X57809 lambda light chain variable region 10.9 10.0 Y00081 IL6 interleukin 6 (interferon, beta 2) 9.2 9.4 D16583 HDC histidine decarboxylase 8.0 9.4 X07730 KLK3 kallikrein 3, (prostate specific antigen) 5.6 8.8 HG311 -HT3287 Homo sapiens clone HH409 unknown 9.5 7.5 M57732 TCFl hepatic nuclear factor (HNF1) 2.0 7.2 U77735 PIM2 pim-2 oncogene 7.1 7.1 U96094 SLN sarcolipin 12.2 6.1 D50640 PDE3B phosphodiesterase 3B, cGMP-inhibited 4.0 5.4 X14008 LYZ lysozyme (renal amyloidosis) 3.0 5.4 M91036 HBG2 hemoglobin, gamma 0 3.4 5.4 X72755 MIG monokine induced by gamma interferon 5.2 5.2 AC000099 GRMS glutamate receptor, metabotropic 8 2.3 4.3 Dl 1428 PMP22 peripheral myelin protein 22 5.0 4.0 M83667 CBBPD CCAAT/enhancer binding protein (CIEBP), delta 4.3 4.0 L19267 DMWD dystrophia myotonica, WD repeat motif 3.0 3.8 M81181 ATPIB2 ATPase, Na+/K+ transporting 3.5 3.8 U79249 Human clone 23839 sequence 3.1 3.7 U49973 FJ10803 hypothetical protein FLJ10803 3.2 3.6 HG870-HT870 GOLGA3 golgi autoantigen, golgin subfamily a, 3 3.5 3.6 X13589 CYP19 cytochrome P450, subfamily XIX 3.0 3.5 AB000464 clone:RES4-24A 2.9 3.5 M96956 TDGF1 teratocarcinoma-derived growth factor 1 2.6 3.5 U31628 ILISRA interleukin 15 receptor, alpha 6.4 3.3 D38128 PTGIR prostaglandin 12 (prostacyclin) receptor (IP) 8.8 3.3 J03507 C7 complement component 7 2.3 3.1 M32011 NCF2 neutrophil cytosolic factor 2 3.5 3.0 X63131 PML promyelocytic leukemia 4.7 3.0 D82326 SLC3AI solute carrier family 3 4.0 3.0 L10343 P13 protease inhibitor 3, skin-derived (SKALP) 2.1 3.0 U89995 FOXEI forkhend box El (thyroid transcription factor 2) 2.6 2.9 M62800 SSAI (52kD, ribonucleoprotein autoantigen SS-A/Ro) 3.1 2.9 AB000584 PLAB prostate differentiation factor 2.4 2.8 U37519 ALDH8 aldehyde dehydrogenase 8 2.2 2.7 D21267 SNAP25 synaptosomal-associated protein, 25kD 2.2 2.7 M25667 GAP43 growth associated protein 43 2.5 2.7 L34357 GATA4 GATA-binding protein 4 2.3 2.7 U43944 MEl malic enzyme 1, NADP(+)-dependent, cytosolic 3.0 2.7 M16937 HOXB7 homeo box B2 2.9 2.6 U27326 FUT3 fucosyltransferase 3 2.6 2.6 -99- Genbank ID Gene Name Unigene Comment Fold 10 ng Fold 40 ng Z23115 BCL2Ll BCL2-like 1 2.2 2.6 HG1877-HT1917 MBP myelin basic protein 2.4 2.6 D 10995 HTR1B 5-hydroxytryptamine (serotonin) receptor lB 2.5 2.6 M91463 SLC2A4 solute carrier family 2 glucose transporter 3.1 2.5 U19878 TMEFF1 transmembrane with EGF and follistatin like 2.9 2.4 U66468 CGRII cell growth regulatory with EF-hand domain 2.2 2.4 U44848 NRF1 nuclear respiratory factor 1 3.5 2.4 U73328 DLX4 distal-less homeobox 4 3.2 2.4 HG4593-HT4998 voltage-gated sodium channel (SCN1A) 2.3 2.4 X78710 MTFI metal-regulatory transcription factor 1 2.7 2.4 X59727 MAPK4 mitogen-activated protein kinase 4 2.3 2.4 103600 ALOX5 arachidonate 5-lipoxygenase 2.2 2.3 U87269 E4Fl E4F transcription factor 1 3.4 2.3 Y10375 PTPNS1 tyrosine phosphatase, non-receptor substrate 1 4.5 2.2 D49958 GPM6A glycoprotein M6A 3.3 2.2 U60062 FEZI fasciculation & elongation protein zeta 1 (zygin I) 3.3 2.2 X14830 CHRNB1 cholinergic receptor, nicotinic, beta polypeptide 1 2.4 2.1 J04076 EGR2 early growth response 2 (Krox-20 homolog) 3.0 2.1 HG2981-HT3127 CD44 CD44 antigen 2.2 2.1 U49187 C60RF32 chromosome 6 open reading frame 32 3.8 2.1 X77744 Homo sapiens for FLJ00032 protein, partial 2.3 2.1 X68285 GK glycerol kinase 2.4 2.0 HG3925-HT4195 SFTPA2 surfactant, pulmonary-associated protein A2 3.9 2.0 M26062 IL2RB interleukin 2 receptor, beta 0.2 0.5 X06182 KIr v-kit oncogene homolog 0.4 0.5 U79251 OPCML opioid-binding protein/cell adhesion molecule-like 0.5 0.5 103764 SERPINEI nexin, plasminogen activator inhibitor type 1 0.5 0.5 X92814 HREV107 similar to rat HREV107 0.3 0.5 L01087 PRKCQ protein kinase C, theta 0.2 0.5 D43772 GRB7 growth factor receptor-bound protein 7 0.2 0.5 X15880 COL6A1 collagen, type VI, alpha 1 0.5 0.5 H03115-HT3291 MBP myelin basic protein 0.4 0.5 X83301 SMA3 SMA3 0.5 0.5 D87469 CELSR2 cadherin, EGF LAG seven-pass G-type receptor 2 0.4 0.5 Ml 1313 A2M alpha-2-macroglobulin 0.4 0.4 X64877 HFL3 H factor (complement)-like 3 0.4 0.4 Z18859 ONAT2 guanine nucleotide binding protein (G protein) 0.4 0.4 D89077 SLA Src-like-adapter 0.4 0.4 L25444 TAF2E TATA box binding protein (TBP)-associated factor 0.2 0.4 M26665 HTN3 histatin 3 0.4 0.4 S69790 WASF3 WAS protein family, member 3 0.4 0.4 U79248 Human clone 23826 sequence 0.4 0.4 L15309 ZNF141 zinc finger protein 141 (clone pHZ-44) 0.3 0.4 LA1 147 HTR6 5-hydroxytryptarnine (serotonin) receptor 6 0.4 0.4 X58431 HOXB6 homeo box B6 0.4 0.4 U50360 CAMK2G CaM kinase II gamma 0.2 0.4 D88152 ACATN acetyl-Coenzyme A transporter 0.4 0.4 U38480 RXRG retinoid X receptor, gamma 0.3 0.4 X16866 CYP2D7AP cytochrome P450, subfamily ID 0.4 0.4 -100- Genbank ID Gene Name Unigene Comment Fold 10 ng Fold 40 ng X70991 NAB2 NGFI-A binding protein 2 (ERGI bp 2) 0.2 0.4 M60830 EV12B ecotropic viral integration site 2B 0.4 0.4 M27492 ILlRI interleukin I receptor, type I 0.4 0.4 Z35093 SURFI surfeit 1 0.4 0.4 D86425 NID2 nidogen 2 0.3 0.3 U59914 MADH6 MAD) homolog 6 0.4 0.3 M18255 PRKCB1 protein kinase C, beta 1 0.4 0.3 AF000234 P2RX4 purinergic receptor P2X 0.3 0.3 577763 NFE2 nuclear factor (erythroid-derived 2), 45kD 0.4 0.3 U78722 ZNF165 zinc finger protein 165 0.3 0.3 L05568 SLC6A4 solute carrier family 6 (serotonin), 0.3 0.3 L31529 SNTBI syntrophin, dystrophin-associated protein Al. 0.3 0.3 U47054 ART3 ADP-ribosyltransferase 3 0.4 0.3 M13955 KRT7 keratin 7 0.4 0.3 D15049 PTPRH protein tyrosine phosphatase, receptor type, H 0.4 0.3 U03486 GJA5 gap junction protein, alpha 5, 4OkD (connexin 40) 0.5 0.3 X06256 ITGA5 integrin, alpha 5 0.4 0.3 U22314 REST REI-silencing transcription factor 0.3 0.3 U51096 CDX2 caudal type homeo box transcription factor 2 0.2 0.2 D31762 KIAA0057 TRAM-like protein 0.4 0.2 M23668 FDXI ferredoxin 1 0.2 0.2 U53476 WNT7A wingless-type MMTV integration site family 0.2 0.2 X57206 ITPKB inositol 1.4,5-trisphosphate 3-kinase B 0.2 0.2 Z31695 INPP5A inositol polyphosphate-5-phosphatase, 4OkD 0.4 0.2 S66793 ARR3 arresting 3, retinal (X-arrestin) 0.2 0.2 U59877 RAB31 RAB31, member RAS oncogene family 0.2 0.2 U53786 EVPL envoplakin 0.2 0.2 S83362 LIFR leukemia inhibitory factor receptor 0.3 0.2 D42038 KIAA0087 KIAAOOB7 gene product 0.3 0.2 HG4333-HT4603 ZNF79 zinc finger protein 79 (pT7) 0.1 0.1 L01406 GHRHR growth hormone releasing hormone receptor 0.4 0.1 Example 3.2.3: Human Leukocyte Response to IL18. The response of human leukocytes (isolated leukophoresis) to respond to IL18 alone or in combination with 1L12 was tested. The ability of an anti-IL18 monoclonal antibody to inhibit the 5 transcriptional response was also tested. Cells were treated as described Example 4.1. RNA was isolated and used to probe Affymetrix Genechips (Hugene, FL). The results are shown in Table 23, which lists 49 transcripts induced by IL18+L12 and reversed by anti-IL18 antibody. Several genes were relevant to the immune system. Many of these genes were also induced by IL18 in KG-i cells. 10 -101- Table 23. Other potential IL18/fL12 markers selected transcripts upregulated four fold or more by IL18+IL12 and reversed by 1252H in a human leukocyte sample as determined using Affymetrix Genechips. Gene Name Unigene Comment Unigene KIAA0001 putative G protein coupled receptor for UDP-glucose Hs.2465 LIMK2 LIM domain kinase 2 Hs.278027 KLAAO196 KIAA0196 gene product Hs.8294 IFNG interferon, gamma Hs.856 POLR2C polymerase (RNA) U polypeptide Hs.79402 DAGI dystroglycan I Hs.76111 TPSBI tryptase beta I Hs.250700 CDR2 cerebellar degeneration-related protein (62kD) Hs.75124 TCF1 2 helix-loop-helix transcription factors 4 Hs.21704 TACTILE T cell activation, increased late expression Hs. 142023 PIP5K2A phospliatidylinositol-4-phosphate 5-kinase Hs.108966 SP3A3 splicing factor 3a. subunit 3, 60kD Hs.77897 SELIL sel-I (suppressor of lin-12, C.elegans)-like Hs. 181300 ILl5 interleukin 15 Hs.168132 BAKI BCL2-antagonist/killer I Hs.93213 SLAM signaling lymphocytic activation molecule Hs.32970 SCYBIlI small inducible cytokine subfamily B (Cys-X-Cys), member II Hs.103982 LIMKI LIM domain kinase I Hs.36566 CAT56 CAT56 protein Hs. 118354 POLRMT polymerase (RNA) mitochondrial (DNA directed) Hs.153880 SCYA4 small inducible cytosine A4/Mip-lb Hs.75703 MIG monokine induced by gamma interferon Hs.77367 SSX3 synovial sarcoma, X breakpoint 3 Hs.178749 TNFRSP6 tumor necrosis factor receptor superfamily, member 6 Hs.82359 MATIA methionine adenosyltransferase I, alpha Hs.323715 KIAA0133 KIAA0133 gene product Hs.57730 FCGBP Fc fragment of IgG binding protein Hs. 111732 ARHD ras homolog gene family, member Hs. 15114 FGFR2 fibroblast growth factor receptor 2 Hs.278581 COL9AI collagen, type IX, alpha I Hs.154850 HPX42B haemopoictic progenitor homeobox Hs.125231 TAL2 T-cell acute lymphocytic leukemia 2 Hs.247978 ESTs Hs.196244 REN renin Hs.3210 POU2P2 POU domain, class 2, transcription factor 2 Hs.i 101 ALOX12 arachidonate 12-lipoxygenase Hs. 1200 ACTN2 actinin, alpha 2 Hs.83672 KLK2 kallikrein 2, prostatic Hs.181350 -102- Gene Name Unigene Comment Unigene RCVI recovering Hs.80539 E2F4 E2F transcription factor 4, p107/pI30-binding Hs.108371 SEMA3F immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3F Hs.32981 BHMT betaine-homocysteine methyltransferase Hs.80756 EVPL envoplakin Hs.25482 BBC3 Bcl-2 binding component 3 Hs.87246 SLIN sarcolipin Hs.15219 RDBP RD RNA-binding protein Hs.106061 MTI H metallothionein 1H Hs.2667 RADS4L RAD54 (S.cerevisiae)-like Hs.66718 MLL3 myeloid/lymphoid or mixed-lineage leukemia Hs.288971 Example 3.2.4: Human whole blood response to 1L18. The response of whole human blood to respond to IL18 alone or in combination with IL12 was tested. The ability of an anti-IL18 monoclonal antibody to inhibit the transcriptional 5 response was also tested. Normal donor blood samples were treated as described in Example 4.1. RNA was isolated and used to probe Affymetrix Genechips (HugeneFL). The results are shown in Table 24 which lists 16 transcripts that were significantly regulated by IL18+IL12 and reversed by anti-IL18 antibody in whole blood samples isolated from two healthy donors. Several genes were relevant to the immune system. We went on to test the response of three of these 10 genes in panel of 10 normal donors using quantitative PCR. The results of this human variability study are shown in Table 25, for interferon gamma; Table 26, CXCL9 and Table 27, CCL8. The results of the variability study indicated that regulation of these transcripts by IL18 in human blood is likely to be a common among humans. 15 Table 24. Other potential IL18/IL12 markers selected from transcripts up-regulated in whole blood isolated from two donors then treated with IL18+IL12. Probe Set ID Title Unigene 202284_s_at cyclin-dependent kinase inhibitor 1A (p21, Cip1) Hs.179665 202531 at interferon regulatory factor 1 Hs.80645 204057_at interferon consensus sequence binding protein 1 Hs.14453 205488_at granzyme A (granzyme 1, cytotoxic T-lymphocyte- Hs.90708 associated serine esterase 3) 206554.x_at SET domain and mariner transposase fusion gene Hs.265855 206817_x_at trinucleotide repeat containing 4 Hs.26047 207509_s at leukocyte-associated Ig-like receptor 2 Hs.43803 209546_s_at apolipoprotein L, 1 Hs.114309 214438_at H2.0-like homeo box 1 (Drosophila) Hs.74870 -103- 214450_at cathepsin W (lymphopain) Hs.87450 216950 s at FcRI b form (AA 1-344) [Homo sapiens], mRNA sequence Hs.382006 217933_s at leucine aminopeptidase 3 Hs.182579 219386_s at B lymphocyte activator macrophage expressed Hs.20450 219956_at UDP-N-acetyl-alpha-D-galactosanine:polypeptide N- Hs. 151678 acetylgalactosaminyltransferase 6 219971_at interleukin 21 receptor Hs.210546 221223_x at cytokine inducible SH2-containing protein Hs.8257 TAble 25. Interferon y performance in ten human blood samples. p<0.05 for inhibition by either antibody IFN Unstimulated Stimulated Anti-IL18 2.5 Anti-IL18 125-21H donor3n 0.001 0.187 0.014 0.026 donor 5n 0.003 0.012 0.006 0.006 donor 9n 0.001 1.250 0.037 0.000 donorlOn 0.002 0.361 0.024 0.002 donorin 0.002 0.339 0.022 0.070 donor2n 0.001 0.032 0.003 0.003 donor4n 0.001 0.082 0.011 0.027 donor6n 0.002 0.076 0.006 0.010 donor7n 0.002 0.049 0.009 0.012 donor8n 0.002 0.049 0.009 0.012 5 Table 26. MIG/CXCL9 performance in ten human blood samples. p<0.05 for inhibition by either antibody. CXCL9 Unstimulated Stimulated Anti-IL18 2.5 Anti-IL18 125-2H donors 0.000 0.170 0.082 0.010 donorlO 0.000 0.015 0.000 0.000 donor2 0.001 0.006 0.001 0.001 donor3 0.000 0.067 0.010 0.006 donor4 0.000 0.023 0.012 0.003 donors 0.000 0.004 0.000 0.000 donor6 0.000 0.070 0.001 0.001 donor7 0.001 0.034 0.001 0.000 donor8 0.001 0.034 0.001 0.000 donor9 0.000 0.035 0.000 0.001 10 -104- Table 27. MCP2/CCL8 performance in ten human blood samples. p<0.05 for inhibition by either antibody. CCL8 Unstimulated Stimulated Anti-1L18 2.5 Anti-IL18 125-2H donors 0.036 8.941 4.054 1.051 donorlO 0.004 0.987 0.009 0.025 donor2 0.036 1.225 0.105 0.057 donor3 0.012 3.923 0.648 . 0.663 donor4 0.021 2.227 0.994 0.630 donor5 0.001 0.005 0.001 0.001 donor6 0.000 0.023 0.002 0.001 donor7 0.001 0.009 0.001 0.001 donor8 0.001 0.009 0.001 0.001 donor9 0.001 2.438 0.003 0.059 5 Example 4: Characterization of anti-IL-18 HuMAb, 2.13(E)mg1 Example 4.1: Human Cytokine Specificity The specificity of 2.13(E)mgl for human IL-18 was evaluated using the BIACORE assay following manufacturer's instructions (see Example 2. 1.B). The 2.13(E)mgl was captured on a biosensor chip and its ability to bind a panel of known human cytokines in solution was 10 determined. As shown in Table 28, 2.13(E)mgl bound recombinant mature human IL-18. However, 2.13(E)mgl did not bind human proIL-18 nor did it bind any of the other 23 human cytokines tested, including the IL-1 family members IL-la and IL-1 Table 28 Biacore Analysis of Cytokine Binding by 2.13(E)mgl and 2.5(E)mgl 15 Soluble rec. human Captured 2.13(E)mg1 Captured 2.5(E)mgl cytokines, (1pM) (25 mg/mL) (25 mg/mL) 2.13(E)mg1 Binding 2.5(E)MG1Binding IFNy IL-la IL-1@ - _ Other cytokinesa IL-18 Pro-IL-18 + Additional cytokines tested for binding included IL-2, IL-3, 11-4, IL-5, IL-6, IL-7, IL-8, IIL-9, IL-10, IL-l , IL-12, IL-13,IL-15, IL-16, IL-17, I-21, TNF, LT, LTalP2, and LTa2o1. 2.13(E)mgl did not bind to any of these cytokines. Cysteine > Alanine mutant BV derived recombinant human IL-18 -105- Example 4.2: Competition with other antibodies to bind human IL-18 5 The ability of several anti-IL-18 antibodies to compete with 2.13(E)mgl for binding to human IL-18 was evaluated using the BIACORE assay following manufacturer's instructions (see Example 2.1.B). Briefly, polyclonal anti-human or anti-mouse antibodies were captured on a biosensor chip. Thereafter, anti-IL-18 antibodies were introduced and captured by the polyclonal anti-human or anti-mouse (only for 125-2H) antibodies immobilized on the biosensor chip 10 described above (primary immobilized antibody). Then, recombinant human IL-18 was introduced and captured by the primary immobilized antibody. Finally, secondary soluble anti IL-18 antibodies were introduced. The assay measured the ability of the secondary soluble anti IL-18 to bind the recombinant IL-18 and compete with the primary antibody. 2.13(E)mgl did not compete with either 2.5(E)mglor IL-18BP Murine anti-huIL-18 monoclonal antibody 125-2H 15 competed with 2.13(E)mgl for binding to human IL-18. Table 29 BIACORE analysis of Antibody Competition for binding to human IL-18 20 soluble 10 Immunobilized Ab Ab 125-2H 2.5(E)mgl 215 444 581 435 2.13(E)mgl 2.3 EL-18BP 125-2H - + - - + + - - + 2.5(E)ngl + - + + - - + + + 215 - + - - + + - - + 4- + - - + + - + 581 + - + + - - + + + 43 + - + + - - + + + 2.13(E)mg - + + + + 2.3 - + - - + + - - + IL-18BP + + + + + + + + + indicates that the primary and the secondary antibodies bind simultaneously 20 - indicates that the secondary antibody cannot bind to the captured IL- 18 The present invention incorporates by reference in their entirety techniques well known in the field of molecular biology. These techniques include, but are not limited to, techniques 25 described in the following publications: Ausubel, F.M. et al. eds., Short Protocols In Molecular Biology (4th Ed. 1999) John Wiley & Sons, NY. (ISBN 0-471-32938-X). -106- Lu and Weiner eds., Cloning and Expression Vectors for Gene Function Analysis (2001) BioTechniques Press. Westborough, MA. 298 pp. (ISBN 1-881299-21-X). Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5). 5 Old, R.W. & S.B. Primrose, Principles of Gene Manipulation: An Introduction To Genetic Engineering (3d Ed. 1985) Blackwell Scientific Publications, Boston. Studies in Microbiology; V.2:409 pp. (ISBN 0-632-01318-4). Sambrook, J. et al. eds., Molecular Cloning: A Laboratory Manual (2d Ed. 1989) Cold Spring Harbor Laboratory Press, NY. Vols. 1-3. (ISBN 0-87969-309-6). 10 Winnacker, E.L. From Genes To Clones: Introduction To Gene Technology (1987) VCH Publishers, NY (translated by Horst Ibelgaufts). 634 pp. (ISBN 0-89573-614-4). -107- 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 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 5,985,615 5,994,619 5,998,209 6,054,487 6,060,283 6,075,181 6,091,001 6,114,598 6,130,364 US patent application publication 20030186374 U.S. Application Serial No. 09/428,082 10 Foreign Patent Documents EP 712 931 EP 850 952 EP 864 585 EP 0 962 531 EP 0 974 600 JP 111,399194 IL 121554 A0 WO 91/10741 WO 91/17271 WO 92/01047 WO 92/02551 WO 92/09690 WO 92/15679 WO 92/18619 WO 92/20791 WO 93/01288 WO 94/02602 WO 96/33735 15 WO 96/34096 WO 97/24441 WO 97/29131 WO 98/16654 WO 98/24893 WO 98/41232 WO 98/50433 WO 99/09063 WO 99/22760 WO 99/25044 WO 99/37772 WO 99/37773 WO 99/45031 WO 99/53049 WO 00/37504 WO 00/09560 WO 00/12555 WO 00/37504 WO 00/56772 WO 01/58956 WO 01/83525 WO 02/72636 20 Other References Adachi a., et al. (1998) Immunity 9:143-150 Akita, K. et al., (1997) J. Biol. Chein. 272, 26595-26603 Azzazy H., and Highsmith W.E., (2002) Clin. Biochem. 35:425-445 Babcock, J.S. et al. (1996) Proc. Natl. Acad. Sci. USA 93:7843-7848 25 Barbas et al. (1991) PNAS 88:7978-7982 Bendele, A., et al (1999) Toxicol Pathol. 27:134-142 Bird et al. (1988) Science 242:423-426 Clackson et al. (1991) Nature 352:624-628 Dinarello, C. et al. (1998) J. Leukoc. Biol. 63:658-654 30 Dinarello, C.A. (1999) Methods 19:121-132 Dinarello, C.A. (1999) J. Allergy Clin. Immunol. 103:11-24; Durocher et al., Nucleic Acids Research 2002, Vol 30, No.2 Fuchs et al. (1991) Bio/Technology 9:1370-1372 Garrad et al. (1991) Bio/Technology 9:1373-1377 35 Gavilondo J.V., and Larrick J.W. (2002) BioTeclniques 29:128-145 Giege, R. and Ducruix, A. Barrett, 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. et al., (1997) Nature 386:619-623 Ghetie, V., et al (1997) Nat. Biotechnol. 15:637-640 -108- Gracie J. A., et al., (2003) Journal of Leukocyte Biology 73, 213-224 Gram et al. (1992) PNAS 89:3576-3580 Green et al. Nature Genetics 7:13-21 (1994) Green and Jakobovits 1. Exp. Med. 188:483-495 (1998) 5 Griffiths et al. (1993) EMBO J12:725-734 Gu, Y. et al., (1997) Science 275:206-209) Hay et al. (1992) Hum Antibod Hybridomas 3:81-85 Harlow and Lane, Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Press, 1990. 10 Hawkins et al. (1992) J Mol Biol 226:889-896 Hezareh, M., et. al., (2001) . Virology,75 (24):12161-12168 Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448 Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137 Hoogenboom H.R., (1997) TIB Tech. 15:62-70 15 Hoogenboom H., and Chames P. (2000) Immunology Today 21:371-378 Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883 Hoshino K., et al (1999) J. Immuiol. 162:5041-5044 Huse et al. (1989) Science 246:1275-1281 Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277. 20 Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131 J6nsson, U., et al. (1991) Biotechniques 11:620-627 Jbnsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26 Kanakaraj P., (1999) J. Exp. Med. 189:1129-1138 Kaufman, R.J. and Sharp, P.A., (1982) Mol. Biol. 159:601-621 25 Kearney et al, J. Immunol. 123, 1979, 1548-1550 Kellermann S. A. and Green L.L. (2002) Current Opinion in Biotechnology 13:593-597 Kim J.K., et al (1999) Eur. J. Immunol. 29:2819-2825 Konishi, K., et al (1997) J. Immnunol. Methods 209:187-191 Kipriyanov, S.M., et al. (1994) Mol. Immunol. 31:1047-1058 30 Kipriyanov, S.M., et al. (1995) Human Antibodies and Hybridomas 6:93-101 Leung, B.P., et al (2001) J. Immunol. 167:2879-2886 Little M. et al (2000) Immunology Today 21:364-370 BioTechniques Press. Westborough, MA. 298 pp. (ISBN 1-881299-21-X). Lund, J. et al., J. Immunology (1991) 147: 2657-2662 -109- McCafferty et al., Nature (1990) 348:552-554 McInnes, I.B. et. al. (2000) Immunology Today 21:312-315; Mendez et al., Nature Genetics 15:146-156 (1997) Mizushima, S. and Nagata, S., (1990) Nucleic acids Research Vol 18, No. 17 5 Nakanishi, K. et al (2001) Ann. Rev. Immunol 19:423-474. 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., et al. (1994) Structure 2:1121-1123 10 Seidman, J.G., Smith, J.A., and K. Struhl eds; Wiley Interscience, N.Y., N.Y. (1990) Sims, J.E., (2002) Current Opin Immunol. 14:117-122 Sugawara, S. et al., (2001) J. Immunol., 167, 6568-6575 Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. 15 Takeda, K., et al. (1998) Immunity 8:383-390 Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295 Tissi L., et al (1999) Infect. Immunol. 67:4545-.50 Trentham, D.E. et al (1977) J. Exp. Med. 146:857-868 Tsutsui, H. et al., (1999) Immunity 11:359-67 20 Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220 Ushio, S., et al. (1996) J. Immunol. 156:42744279 Ward et al., (1989) Nature 341:544-546 Wei, X.Q., et al (2000) J. Immunol. 166:517-521 Winnacker, E.L. From Genes To Clones: Introduction To Gene Technoloy (1987) VCH 25 Publishers, NY (translated by Horst Ibelgaufts). 634 pp. (ISBN 0-89573-614-4). Although a number of embodiments and features have been described above, it will be understood by those skilled in the art that modifications and variations of the described embodiments and features may be made without departing from the present disclosure or the 30 invention as defined in the appended claims. Each of the publications mentioned herein is incorporated by reference. -110-

Claims (49)

1. An isolated monoclonal antibody, or antigen binding portion thereof, capable of binding human IL-18, wherein the antibody, or antigen binding portion thereof, comprises: a heavy chain variable region comprising CDR1, CDR2, and CDR3 sequences, and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, wherein the heavy chain variable region CDR3 sequence comprises an amino acid sequence of residues 99-110 of SEQ ID NO:6, or an amino acid sequence of residues 99-110 of SEQ ID NO:6 containing conservative amino acid substitutions.
2. The antibody, or antigen binding portion thereof, of claim 1, wherein the light chain variable region CDR3 sequence comprises an amino acid sequence of residues 89 98 of SEQ ID NO:7 or an amino acid sequence of residues 89-98 of SEQ ID NO:7 containing conservative amino acid substitutions.
3. The antibody, or antigen binding portion thereof, of claim 1 or 2, wherein the heavy chain variable region CDR2 sequence comprises an amino acid sequence of residues 50-66 of SEQ ID NO:6 or an amino acid sequence of residues 50-66 of SEQ ID NO:6 containing conservative amino acid substitutions.
4. The antibody, or antigen binding portion thereof, of any one of claims 1-3, wherein the light chain variable region CDR2 sequence comprises an amino acid sequence of residues 50-56 of SEQ ID NO:7 or an amino acid sequence of residues 50-56 of SEQ ID NO:7 containing conservative amino acid substitutions.
5. The antibody, or antigen binding portion thereof, of any one of claims 1-4, wherein the heavy chain variable region CDR1 sequence comprises an amino acid sequence of residues 31-35 of SEQ ID NO:6 or an amino acid sequence of residues
31-35 of SEQ ID NO:6 containing conservative amino acid substitutions. 6624370 112 6. The antibody, or antigen binding portion thereof, of any one of claims 1-5, wherein the light chain variable region CDR1 sequence comprises an amino acid sequence of residues 24-34 of SEQ ID NO:7 or an amino acid sequence of residues 24-34 of SEQ ID NO:7 containing conservative amino acid substitutions. 7. An isolated monoclonal antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, further comprises a heavy chain variable region CDR1 comprising residues 31-35 of SEQ ID NO:6, a heavy chain variable region CDN2 comprising residues 50-66 of SEQ ID NO:6, a light chain variable region CDR1 comprising residues 24-34 of SEQ ID NO:7, a light chain variable region CDR2 comprising residues 50-56 of SEQ ID NO:7, and a light chain variable region CDR3 comprising residues 89-98 of SEQ ID NO:7. 8. An isolated monoclonal antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, comprises a heavy chain variable region (VH) comprising SEQ ID NO:6. 9. An isolated monoclonal antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, comprises a light chain variable region (VL) comprising SEQ ID NO:7. 10. An isolated antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, comprises: an Ig constant heavy region comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2 and SEQ ID NO: 3; an Ig constant light region comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4 and SEQ ID NO: 5; an Ig variable heavy region comprising the amino acid sequence of SEQ ID NO:6; and an Ig variable light region comprising the amino acid sequence of SEQ ID NO:7. 11. An isolated antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, comprises: 6624370 113 an Ig constant heavy region comprising the amino acid sequence of SEQ ID NO: 3: an Ig constant light region comprising the amino acid sequence of SEQ ID NO:4; an Ig variable heavy region comprising the amino acid sequence of SEQ ID NO:6; and an Ig variable light region comprising the amino acid sequence of SEQ ID NO:7. 12. An isolated antibody, or antigen binding portion thereof, according to claim 1, wherein said antibody, or antigen binding portion thereof, comprises: an Ig variable heavy region comprising the amino acid sequence of SEQ ID NO:6; and an Ig variable light region comprising the amino acid sequence of SEQ ID NO:7. 13. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-12, wherein said antibody or antigen binding portion thereof binds human IL-18 and is not capable of competing with 125-2H for binding human IL-18. 14. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-12, wherein said antibody or antigen binding portion thereof binds human IL-18 and is not capable of competing with IL-18BP for binding human IL-18. 15. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-9 and 12-14, further comprising a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain, a human IgG1 constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgE constant domain, and a human IgA constant domain. 16. The isolated antibody, or antigen binding portion thereof, according to claim 15, wherein said heavy chain immunoglobulin constant domain is a human IgG1 constant domain. 6624370 114 17. The isolated antibody, or antigen binding portion thereof, according to claim 16, wherein said human IgG1 constant domain comprises the amino acid sequence selected from the group consisting of SEQ ID NO:2 and SEQ ID NO:3. 18. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-9 and 12-14, further comprising a light chain immunoglobulin constant domain selected from the group consisting of a human Ig kappa constant domain and a human Ig lambda constant domain. 19. The isolated antibody, or antigen binding portion thereof, according to claim 18, wherein said light chain immunoglobulin constant region domain is a human Ig kappa constant domain comprising the amino acid sequence SEQ ID NO:4. 20. The isolated antibody, or antigen binding portion thereof, according to claim 18, wherein said light chain immunoglobulin constant domain is a human Ig lambda constant domain comprising the amino acid sequence SEQ ID NO:5. 21. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-20, wherein said antibody, or antigen binding portion thereof, is recombinant. 22. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-21, wherein said antibody, or antigen binding portion thereof, is glycosylated. 23. The isolated antibody, or antigen binding portion thereof, according to any one of claims 1-22, wherein said antibody or antigen binding portion thereof is selected from the group consisting of an immunoglobulin molecule, an scFv, a monoclonal antibody, a human antibody, a chimeric antibody, a humanized antibody, a Fab fragment, an Fab' fragment, an F(ab')2, an Fv, and a disulfide linked Fv. 24. The isolated antibody, or antigen binding portion thereof, according to claim 23, wherein said antibody or antigen binding portion thereof is a human antibody. 6624370 115 25. The antibody, or antigen binding portion thereof, of any one of claims 1-24 wherein said antibody or antigen binding portion thereof is capable of neutralizing human IL-18. 26. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said IL-18 is selected from the group consisting of pro-human IL 18, mature-human IL-18, and truncated-human IL-18. 27. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, diminishes the ability of human IL-18 to bind to its receptor. 28. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, diminishes the ability of pro-human IL-18, mature-human IL-18, or truncated human IL-18 to bind to its receptor. 29. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, is capable of reducing one or more of human IL-18 biological activities selected from the group consisting of Th1 modulation, Th2 modulation, NK modulation, neutrophil modulation, monocyte-macrophage lineage modulation, eosinophil modulation, B cell modulation, cytokine modulation, chemokine modulation, adhesion molecule modulation, and cell recruitment modulation. 30. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, has a dissociation constant (KD) selected from the group consisting of about 10- 7 M; about 10~8 M; about 10-9 M; about 10-1 M; about 10-" M; about 10-12 M; and about 10-13 M. 31. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, has 6624370 116 an on-rate selected from the group consisting of about 10 2 M-ls-; about 10 3 M's'; about 10 4 M-Is; about 10"M-ls- 1 ; and about 10 6 Mls'.
32. The isolated neutralizing antibody, or antigen binding portion thereof, according to claim 25, wherein said neutralizing antibody, or antigen binding portion thereof, has an off-rate selected from the group consisting of about 10 3 S 1 ; about 10-s1; about 10 5 s 1 ; and about 10-6s1.
33. A labeled antibody, or antigen binding portion thereof, comprising the antibody, or antigen binding portion thereof, according to any one of claims 1-32, wherein said antibody or antigen binding portion thereof is conjugated to a detectable label.
34. The labeled antibody, or antigen binding portion thereof, according to claim 33, wherein 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.
35. The labeled antibody, or antigen binding portion thereof, according to claim 33, wherein said label is a radiolabel selected from the group consisting of 3 H, 1 4 C, 3s, 90 Y, **Tc, 111 n, 1251 3 177 Lu, 166 Ho, and 1 3 Sm.
36. A conjugate antibody, or antigen binding portion thereof, comprising the antibody, or antigen binding portion thereof, according to any one of claims 1-32, wherein said antibody or antigen binding portion thereof is conjugated to a therapeutic or cytotoxic agent.
37. The conjugate antibody, or antigen binding portion thereof, according to claim 36, wherein said therapeutic or cytotoxic agent is selected from the group consisting of an anti-metabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, a toxin, and an apoptotic agent. 6624370 117
38. An isolated nucleic acid that encodes the antibody, or antigen binding portion thereof, of any one of claims 1-32.
39. A vector comprising the isolated nucleic acid according to claim 38.
40. The vector according to claim 39, wherein said vector is selected from the group consisting of pcDNA, pTT, pTT3, pEFBOS, pBV, pJV, and pBJ.
41. An isolated recombinant host cell comprising the vector according to claim 39 or 40.
42. The recombinant host cell according to claim 41, wherein said host cell is a prokaryotic cell.
43. The recombinant host cell according to claim 42, wherein said host cell is Ecoli.
44. The recombinant host cell according to claim 41, wherein said host cell is an eukaryotic cell.
45. The recombinant host cell according to claim 44, wherein said eukaryotic cell is selected from the group consisting of an animal cell, a plant cell and a fungal cell.
46. The recombinant host cell according to claim 45, wherein said eukaryotic cell is an animal cell selected from the group consisting of: a mammalian cell, an avian cell, and an insect cell.
47. The recombinant host cell according to claim 46, wherein said animal cell is a CHO cell.
48. The recombinant host cell according to claim 46, wherein said host cell is COS.
49. The recombinant host cell according to claim 45, wherein said eukaryotic cell is Saccharomyces cerevisiae. 6624370 118
50. The recombinant host cell according to claim 46, wherein said animal cell is an insect Sf9 cell.
51. A method of producing an antibody, or antigen binding portion thereof, that binds human IL-18, the method comprising the step of culturing the host cell according to claim 41 in a culture medium under conditions sufficient to produce an antibody, or antigen binding portion thereof, that binds human IL-18.
52. A pharmaceutical composition comprising the antibody, or antigen binding portion thereof, according to any one of claims 1-32, and a pharmaceutically acceptable carrier.
53. The pharmaceutical composition according to claim 52, which further comprises at least one additional therapeutic agent for treating a disorder in which IL-18 activity is detrimental.
54. The pharmaceutical composition according to claim 53, 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; methotrexate; corticosteroids; cyclosporin; rapamycin; FK506; and non-steroidal anti-inflammatory agents.
55. The pharmaceutical composition according to claim 52 for treating a disorder selected from the group consisting of rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scieroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, 6624370 119 uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type 11, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, cornnron varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing, alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonilis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/ polymyositis associated lung disease, Sjogren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-i autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease 6624370 120 associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary scierosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, Lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjogren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo, acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders, depression, schizophrenia, Th2 Type and Th1 Type mediated diseases, acute and chronic pain, and cancer.
56. A crystallized antibody, or antigen binding portion thereof, comprising the antibody, or antigen binding portion thereof, of any one of claims 1-32, wherein said antibody, or antigen binding portion thereof, exists as a crystal.
57. The crystallized antibody, or antigen binding portion thereof, according to claim 56, wherein said crystal is a carrier-free pharmaceutical controlled release crystal.
58. The crystallized antibody, or antigen binding portion thereof, according to claim 56, wherein said crystallized antibody, or antigen binding portion thereof, has a greater half life in vivo than the soluble counterpart of said antibody or antigen binding portion thereof. 6624370 121
59. The crystallized antibody, or antigen binding portion thereof, according to claim 56, wherein said crystallized antibody, or antigen binding portion thereof, retains biological activity.
60. A composition for the release of an antibody, or antigen binding portion thereof, said composition comprising (a) a formulation, wherein said formulation comprises a crystallized antibody or antigen binding portion thereof, according to any one of claims 56-59, and an ingredient; and (b) at least one polymeric carrier.
61. The composition according to claim 60, wherein said polymeric carrier is a polymer selected from one or more of the group consisting of: poly acrylic acid, poly cyanoaciylates, poly amino acids, poly anhydrides, poly depsipeptide, poly esters, poly lactic acid, poly lactic-co-glycolic acid or PLGA, poly b-hydroxybutryate, poly caprolactone, poly dioxanone; poly ethylene glycol, poly hydroxypropyl methaciylamide, poly organo phosphazene, poly ortho esters, poly vinyl alcohol, poly vinylpyrrolidone, maleic anhydride- alkyl vinyl ether copolymers, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycaminoglycans, sulfated polysaccharides, blends and copolymers thereof.
62. The composition according to claim 60, wherein said ingredient is selected from the group consisting of: albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl cyclodextrin, methoxypolyethylene glycol and polyethylene glycol.
63. Use of the antibody, or antigen binding portion thereof, of any one of claims 1-32 in the preparation of a medicament for reducing human IL-18 activity.
64. Use of the antibody, or antigen binding portion thereof, of any one of claims 1-32 in the preparation of a medicament for reducing human IL-18 activity in a human subject suffering from a disorder in which IL-18 activity is detrimental. 6624370 122
65. Use of the antibody, or antigen binding portion thereof, of any one of claims 1-32 in the preparation of a medicament for treating a subject for a disease or a disorder in which IL-18 activity is detrimental.
66. The use of claim 65, wherein said 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, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawaski's disease, Grave's disease, nephritic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoelein pupurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type 1l, Schmidt's syndrome, adult respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthopathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthopathy, entereopathic synovitis, Chlamydia, yersinia and salmonella associated arthopathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmunehepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency, dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective 6624370 123 tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interatitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyosititis/polymyositis associated lung disease, Sjorgen's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, radiation fibrosis, bronchiolitis oblierans, chronic eosinophilic pneumonia, lymphocytic infilitrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis, type-2 autoimmune hepatitis, autoimmune mediated hypoglycaemia, type B insulin resistance with scanthosis nigricans, hypoarathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, Lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis, sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritia nodosa, acute rheumatoid fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjorgen's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goltrous autoimmune hypothyroidism, atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uvelitis, primary vasculitis, vitiligo, acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol induced livery injury, choleostatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatititis, allergy and asthma, group B streptococci infection, mental disorders, TH2 Type and TH1 Type mediated diseases, acute and chronic pain, and cancer.
67. The use according to claim 65, wherein said disorder is asthma. 6624370 124
68. The use according to claim 65, wherein said disorder is systemic lupus erythematosus (SLE).
69. The use according to claim 65, wherein said disorder is osteoarthritis.
70. The use according to claim 65, wherein said disorder is chronic obstructive pulmonary disease.
71. The use according to claim 65, wherein said disorder is psoriasis.
72. The use according to claim 65, wherein said disorder is psoriatic arthritis.
73. Use of an antibody, or antigen binding portion thereof, of any one of claims 1-32 and a second agent in the preparation of a medicament for treating a patient for a disease or a disorder in which IL-18 is detrimental, wherein the medicament is formulated for administering the antibody, or antigen binding portion thereof, before, concurrent, or after the administration of a second agent, and wherein the second agent is selected from the group consisting of an antibody, or fragment thereof, capable of binding human IL-12; methotrexate; an antibody, or fragment thereof, capable of binding human TNF; corticosteroids; cyclosporine; rapamycin; FK506; and non-steroidal anti-inflammatory agents.
74. A method for reducing human IL-18 activity in vitro, the method comprising the step of contacting human IL-18 with the antibody, or antigen binding portion thereof, of any one of claims 1-32, such that human IL-18 activity is reduced. Dated 21 August, 2012 Abbott Laboratories Patent Attorneys for the Applicant/Nominated Person SPRUSON & FERGUSON 6624370 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011 2011224023 13 Sep 2011
AU2011224023A 2003-11-12 2011-09-13 IL-18 binding proteins Ceased AU2011224023C1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2011224023A AU2011224023C1 (en) 2003-11-12 2011-09-13 IL-18 binding proteins
AU2012261666A AU2012261666A1 (en) 2003-11-12 2012-12-11 IL-18 binding proteins

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10/706,689 2003-11-12
AU2004290073A AU2004290073B2 (en) 2003-11-12 2004-11-12 IL-18 binding proteins
AU2011224023A AU2011224023C1 (en) 2003-11-12 2011-09-13 IL-18 binding proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2004290073A Division AU2004290073B2 (en) 2003-11-12 2004-11-12 IL-18 binding proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2012261666A Division AU2012261666A1 (en) 2003-11-12 2012-12-11 IL-18 binding proteins

Publications (3)

Publication Number Publication Date
AU2011224023A1 AU2011224023A1 (en) 2011-10-06
AU2011224023B2 true AU2011224023B2 (en) 2012-09-13
AU2011224023C1 AU2011224023C1 (en) 2013-08-29

Family

ID=45439601

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2011224023A Ceased AU2011224023C1 (en) 2003-11-12 2011-09-13 IL-18 binding proteins
AU2012261666A Abandoned AU2012261666A1 (en) 2003-11-12 2012-12-11 IL-18 binding proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2012261666A Abandoned AU2012261666A1 (en) 2003-11-12 2012-12-11 IL-18 binding proteins

Country Status (1)

Country Link
AU (2) AU2011224023C1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015032932A1 (en) 2013-09-05 2015-03-12 Ab2 Bio Sa Il-18 binding protein (il-18bp) in inflammatory diseases
MX2017010919A (en) 2015-03-05 2017-12-07 Ab2 Bio Sa Il-18 binding protein (il-18bp) and antibodies in inflammatory diseases.

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0974600A2 (en) * 1998-06-24 2000-01-26 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Anti-interleukin-18 antibody
WO2000056771A1 (en) * 1999-03-19 2000-09-28 Smithkline Beecham Corporation Recombinant il-18 antagonists useful in treatment of il-18 mediated disorders
WO2001058956A2 (en) * 2000-02-10 2001-08-16 Abbott Laboratories Antibodies that bind human interleukin-18 and methods of making and using

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0974600A2 (en) * 1998-06-24 2000-01-26 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Anti-interleukin-18 antibody
WO2000056771A1 (en) * 1999-03-19 2000-09-28 Smithkline Beecham Corporation Recombinant il-18 antagonists useful in treatment of il-18 mediated disorders
WO2001058956A2 (en) * 2000-02-10 2001-08-16 Abbott Laboratories Antibodies that bind human interleukin-18 and methods of making and using

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HOLMES, S. et al, Hybridoma, 2000, 19(5), 363-367 *

Also Published As

Publication number Publication date
AU2011224023A1 (en) 2011-10-06
AU2012261666A1 (en) 2013-01-10
AU2011224023C1 (en) 2013-08-29

Similar Documents

Publication Publication Date Title
AU2004290073B2 (en) IL-18 binding proteins
US7968684B2 (en) IL-18 binding proteins
AU2006265932B2 (en) IL-12/p40 binding proteins
AU2012238334A1 (en) IL-12/p40 binding proteins
AU2011224023B2 (en) IL-18 binding proteins
MXPA06005469A (en) Il-18 binding proteins

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 21 DEC 2012 .

PC Assignment registered

Owner name: ABBVIE INC.

Free format text: FORMER OWNER WAS: ABBOTT LABORATORIES

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 21 DEC 2012 AND 25 FEB 2013

MK14 Patent ceased section 143(a) (annual fees not paid) or expired